WO2017214339A1 - Anti-b7-h3 antibodies and antibody drug conjugates - Google Patents

Anti-b7-h3 antibodies and antibody drug conjugates Download PDF

Info

Publication number
WO2017214339A1
WO2017214339A1 PCT/US2017/036449 US2017036449W WO2017214339A1 WO 2017214339 A1 WO2017214339 A1 WO 2017214339A1 US 2017036449 W US2017036449 W US 2017036449W WO 2017214339 A1 WO2017214339 A1 WO 2017214339A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
antibody
amino acid
acid sequence
methyl
Prior art date
Application number
PCT/US2017/036449
Other languages
French (fr)
Other versions
WO2017214339A4 (en
Inventor
Lorenzo Benatuil
Milan Bruncko
Debra Chao
Kamel IZERADJENE
Andrew S. Judd
Andrew C. PHILLIPS
Andrew J. Souers
Archana THAKUR
Original Assignee
Abbvie Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Abbvie Inc. filed Critical Abbvie Inc.
Priority to BR112018075649-0A priority Critical patent/BR112018075649A2/en
Priority to CN201780048428.5A priority patent/CN109641962A/en
Priority to EP17732658.4A priority patent/EP3469000A1/en
Priority to AU2017279554A priority patent/AU2017279554A1/en
Priority to US16/308,742 priority patent/US20200338209A1/en
Priority to JP2018564199A priority patent/JP2019521973A/en
Priority to CA3027103A priority patent/CA3027103A1/en
Priority to MX2018015285A priority patent/MX2018015285A/en
Publication of WO2017214339A1 publication Critical patent/WO2017214339A1/en
Publication of WO2017214339A4 publication Critical patent/WO2017214339A4/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • B7-H3 The B7 homology 3 protein (B7-H3) (also known as CD276 and B7RP-2, and referred to herein as“B7-H3”) is a type I transmembrane glycoprotein of the immunoglobulin superfamily. Human B7-H3 contains a putative signal peptide, V-like and C-like Ig domains, a transmembrane region and a cytoplasmic domain.
  • B7-H3 isoforms having either a single IgV-IgC-like domain (2IgB7-H3 isoform) or a IgV-IgC-IgV-IgC-like domain (4IgB7-H3 isoform) containing several conserved cysteine residues.
  • the predominant B7-H3 isoform in human tissues and cell lines is the 4IgB7-H3 isoform (Steinberger et al., J. Immunol. 172(4): 2352-9 (2004)).
  • B7-H3 has been reported as having both co-stimulatory and co-inhibitory signaling functions (see, e.g., Chapoval et al., Nat. Immunol.2: 269-74 (2001); Suh et al., Nat. Immunol.4: 899-906 (2003); Prasad et al., J. Immunol.173: 2500-6 (2004); and Wang et al., Eur. J. Immunol.35: 428-38 (2005)).
  • B7-H3 co-stimulatory function since B7-H3 was able to increase proliferation of cytotoxic T-lymphocytes (CTLs) and upregulate interferon gamma (IFN- ⁇ ) production in the presence of anti-CD3 antibody to mimic the T cell receptor signal
  • murine B7-H3 has been found to bind to the triggering receptor expressed on myeloid cells (TREM-) like transcript 2 (TLT-2), a modulator of adaptive an innate immunity cellular responses. Binding of murine B7-H3 to TLT-2 on CD8 + T-cells enhances T-cell effector functions such as proliferation, cytotoxicity and cytokine production (Hashiguchi et al., Proc. Nat'l. Acad. Sci. U.S.A. 105(30): 10495-500 (2008)).
  • B7-H3 is not constitutively expressed in many immune cells ⁇ e.g., natural killer (NK) cells, T- cells, and antigen-presenting cells (APCs)); however, its expression can be induced. Further, the expression of B7-H3 is not restricted to immune cells.
  • B7-H3 transcripts are expressed in a variety of human tissues including colon, heart, liver, placenta, prostate, small intestine, testis, and uterus, as well as osteoblasts, fibroblasts, epithelial cells, and other cells of non-lymphoid lineage, potentially indicating immunological and non-immunological functions (Nygren et al. Front Biosci. 3:989-93 (2011)). However, protein expression in normal tissue is typically maintained at a low level and thus, may be subject to post-transcriptional regulation.
  • B7-H3 is also expressed in a variety of human cancers, including prostate cancer, clear cell renal cell carcinoma, glioma, melanoma, lung cancer, non-small cell lung cancer (NSCLC), small cell lung cancer, pancreatic cancer, gastric cancer, acute myeloid leukemia (AML), non-Hodgkin's lymphoma (NHL), ovarian cancer, colorectal cancer, colon cancer, renal cancer, hepatocellular carcinoma, kidney cancer, head and neck cancer, hypopharyngeal squamous cell carcinoma, glioblastoma, neuroblastoma, breast cancer, endometrial cancer, and urothelial cell carcinoma.
  • human cancers including prostate cancer, clear cell renal cell carcinoma, glioma, melanoma, lung cancer, non-small cell lung cancer (NSCLC), small cell lung cancer, pancreatic cancer, gastric cancer, acute myeloid leukemia (AML), non-Hodgkin's lymphoma (NHL),
  • B7-H3 Although the role of B7-H3 in cancer cells is unclear, its expression may orchestrate signaling events that may protect cancer cells from innate and adaptive immune responses. For example, B7-H3 is overexpressed in high-grade prostatic intraepithelial neoplasia and adenocarcinomas of the prostate, and high expression levels of B7-H3 in these cancerous cells is associated with an increased risk of cancer progression after surgery (Roth et al. Cancer Res. 67(16): 7893-900 (2007)). Further, tumor B7-H3 expression in NSCLC inversely correlated with the number of tumor-infiltrating lymphocytes and significantly correlated with lymph node metastasis (Sun et al.
  • B7-H3 may also play an important role in T-cell-mediated antitumor responses in a context dependent manner.
  • gastric cancer tumor cell expression of B7-H3 positively correlated with survival time, infiltration depth, and tissue type (Wu et al., World J. Gastroenterol. 12(3): 457-9 (2006)).
  • high expression of B7-H3 in pancreatic tumor cells was associated with patient survival after surgical resection and significantly correlated with the number of tumor-infiltrating CD8 + T-cells (Loos et al, BMC Cancer 9:463 (2009).
  • Antibody drug conjugates represent a relatively new class of therapeutics comprising an antibody conjugated to a cytotoxic drug via a chemical linker.
  • the therapeutic concept of ADCs is to combine binding capabilities of an antibody with a drug, where the antibody is used to deliver the drug to a tumor cell by means of binding to a target surface antigen, including target surface antigens that are overexpressed in the tumor cells.
  • a target surface antigen including target surface antigens that are overexpressed in the tumor cells.
  • the present invention provides for antibodies and antibody drug conjugates (ADCs) that specifically bind to human B7-H3.
  • ADCs antibody drug conjugates
  • the present invention provides novel ADCs that can selectively deliver Bcl-xL inhibitors to target cancer cells, e.g., B7-H3 expressing cells.
  • the present invention provide an anti-B7H3 antibody, or antigen binding portion thereof, that binds to human B7-H3 (hB7-H3), wherein the anti-B7H3 antibody, or antigen binding portion thereof, comprises a heavy chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 12 and a light chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 15.
  • the anti-B7H3 antibody, or antigen binding portion thereof comprises a heavy chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 140 and a light chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 7.
  • the anti-B7H3 antibody, or antigen binding portion thereof comprises a heavy chain variable region comprising a CDR1 having the amino acid sequence of SEQ ID NO: 10 and a light chain variable region comprising a CDR1 having the amino acid sequence of either SEQ ID NO: 136 or 138.
  • the present invention provides an anti-B7H3 antibody antibody, or antigen binding portion thereof, that binds to human B7-H3, wherein the anti-B7H3 antibody, or antigen binding portion thereof, comprises a heavy chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 35 and a light chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 39.
  • the anti-B7H3 antibody, or antigen binding portion thereof comprises a heavy chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 34, and a light chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 38.
  • the anti-B7H3 antibody, or antigen binding portion thereof comprises a heavy chain variable region comprising a CDR1 having the amino acid sequence of SEQ ID NO: 33 and a light chain variable region comprising a CDR1 having the amino acid sequence of either SEQ ID NO: 37.
  • the anti-B7H3 antibody, or antigen binding portion thereof is an IgG isotype.
  • the anti-B7H3 antibody, or antigen binding portion thereof is an IgG1 or an IgG4 isotype.
  • the anti-B7H3 antibody, or antigen binding portion thereof has a K D of 1.5 x 10 -8 or less as determined by surface plasmon resonance.
  • the present invention provides an anti-B7H3 antibody, or antigen-binding portion thereof, that binds to hB7-H3, said anti-B7H3 antibody, or antigen-binding portion thereof, comprising a heavy chain variable region comprising either a CDR set of SEQ ID NOs: 10, 11, and 12, and a light chain variable region comprising a CDR set of SEQ ID NOs: 14, 7, and 15; or a heavy chain variable region comprising a CDR set of SEQ ID NOs: 33, 35, and 35, and a light chain variable region comprising a CDR set of SEQ ID NOs: 37, 38, and 39.
  • the anti-B7H3 antibody, or antigen binding portion thereof is humanized. In one embodiment, the anti-B7H3 antibody, or antigen binding portion thereof, further comprises a human acceptor framework.
  • the human acceptor framework comprises an amino acid sequence selected from the group consisting of SEQ ID Nos: 155, 156, 164, 165, 166, and 167. In one embodiment, the human acceptor framework comprises at least one framework region amino acid substitution. In one embodiment, the amino acid sequence of the framework is at least 65% identical to the sequence of the human acceptor framework and comprises at least 70 amino acid residues identical to the human acceptor framework.
  • the amino acid sequence of the framework is at least 85% identical, 90% identical, 95% identical, 96% identical, 97% identical, 98% identical, or 99% identical to the sequence of the human acceptor framework and comprises at least 70, at least 75, at least 80, or at least 85 amino acid residues identical to the human acceptor framework.
  • the human acceptor framework comprises at least one framework region amino acid substitution at a key residue, said key residue selected from the group consisting of: a residue adjacent to a CDR; a glycosylation site residue; a rare residue; a residue capable of interacting with human B7-H3; a residue capable of interacting with a CDR; a canonical residue; a contact residue between heavy chain variable region and light chain variable region; a residue within a Vernier zone; and a residue in a region that overlaps between a Chothia-defined variable heavy chain CDR1 and a Kabat-defined first heavy chain framework.
  • the key residue is selected from the group consisting of 48H, 67H, 69H, 71H, 73H, 94H, and 2L (H refers to the heavy chain; L refers to the light chain; amino acid residues in reference to the Kabat numbering system).
  • the key residue substitution is in the variable heavy chain region and is selected from the group consisting of M48I, V67A, I69L, A71V, K73R, and R94G. In one embodiment, the key residue substitution is in the variable light chain region and is I2V.
  • the present invention provides an anti-B7H3 antibody, or antigen-binding portion thereof, that binds to hB7-H3 comprising a heavy chain variable region comprising a CDR set of SEQ ID NOs: 25, 26, and 27, and a light chain variable region comprising a CDR set of SEQ ID NOs: 29, 30, and 31.
  • the anti-B7H3 antibody, or antigen binding portion thereof is humanized.
  • the antibody, or antigen binding portion thereof further comprises a human acceptor framework.
  • the human acceptor framework comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 155 to 158.
  • the present invention provides an anti-B7H3 antibody, or antigen-binding portion thereof, that binds to hB7-H3 comprising a heavy chain variable region comprising a CDR set of SEQ ID NOs: 33, 35, and 35, and a light chain variable region comprising a CDR set of SEQ ID NOs: 37, 38, and 39.
  • the anti-B7H3 antibody, or antigen binding portion thereof is humanized.
  • the anti-B7H3 antibody, or antigen binding portion thereof further comprises a human acceptor framework.
  • human acceptor framework comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 156, 158, 166 and 167.
  • the human acceptor framework comprises at least one framework region amino acid substitution.
  • the amino acid sequence of the framework is at least 65% identical to the sequence of the human acceptor framework and comprises at least 70 amino acid residues identical to the human acceptor framework.
  • the amino acid sequence of the framework is at least 85% identical, 90% identical, 95% identical, 96% identical, 97% identical, 98% identical, or 99% identical to the sequence of the human acceptor framework and comprises at least 70, at least 75, at least 80, or at least 85 amino acid residues identical to the human acceptor framework.
  • the human acceptor framework comprises at least one framework region amino acid substitution at a key residue, said key residue selected from the group consisting of: a residue adjacent to a CDR; a glycosylation site residue; a rare residue; a residue capable of interacting with human B7-H3; a residue capable of interacting with a CDR; a canonical residue; a contact residue between heavy chain variable region and light chain variable region; a residue within a Vernier zone; and a residue in a region that overlaps between a Chothia-defined variable heavy chain CDR1 and a Kabat-defined first heavy chain framework.
  • the key residue is selected from the group consisting of 69H, 46L, 47L, 64L, and 71L (H refers to the heavy chain; L refers to the light chain; amino acid residues in reference to the Kabat numbering system).
  • the key residue substitution is in the variable heavy chain region and is L69I.
  • the key residue substitution is in the variable light chain region and is selected from the group consisting of L46P, L47W, G64V, and F71H.
  • the present invention provides an anti-hB7-H3 antibody, or antigen- binding portion thereof, comprising a heavy chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 10, a heavy chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 140, a heavy chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 12, a light chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 136 or 138, a light chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 7, and a light chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 15.
  • the present invention provides an anti-hB7-H3 antibody, or antigen- binding portion thereof comprising a heavy chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 33, a heavy chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 34, a heavy chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 35, a light chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 37, a light chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 38, and a light chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 39.
  • the present invention provides an anti-hB7-H3 antibody, or antigen- binding portion thereof, comprising a heavy chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 139 and a light chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 135.
  • the present invention provides an anti-hB7-H3 antibody, or antigen- binding portion thereof, comprising a heavy chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 139, and/or a light chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 135.
  • the present invention provides an anti-hB7-H3 antibody, or antigen- binding portion thereof, comprising a heavy chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 139 and a light chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 137.
  • the present invention provides an anti-hB7-H3 antibody, or antigen- binding portion thereof, comprising a heavy chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 139, and/or a light chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 137.
  • the present invention provides an anti-hB7-H3 antibody, or antigen- binding portion thereof, comprising a heavy chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 147 and a light chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 144.
  • the present invention provides an anti-hB7-H3 antibody, or antigen- binding portion thereof, comprising a heavy chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 147, and/or a light chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 144.
  • the present invention provide an anti-B7H3 antibody that binds to human B7- H3 (hB7-H3), wherein the anti-B7H3 antibody comprises a heavy chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 12 and a light chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 15.
  • the anti-B7H3 antibody comprises a heavy chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 140 and a light chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 7.
  • the anti-B7H3 antibody comprises a heavy chain variable region comprising a CDR1 having the amino acid sequence of SEQ ID NO: 10 and a light chain variable region comprising a CDR1 having the amino acid sequence of either SEQ ID NO: 136 or 138.
  • the present invention provides an anti-B7H3 antibody antibody that binds to human B7-H3, wherein the anti-B7H3 antibody comprises a heavy chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 35 and a light chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 39.
  • the anti-B7H3 antibody comprises a heavy chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 34, and a light chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 38.
  • the anti-B7H3 antibody comprises a heavy chain variable region comprising a CDR1 having the amino acid sequence of SEQ ID NO: 33 and a light chain variable region comprising a CDR1 having the amino acid sequence of either SEQ ID NO: 37.
  • the anti-B7H3 antibody is an IgG isotype.
  • the anti-B7H3 antibody is an IgG1 or an IgG4 isotype.
  • the anti-B7H3 antibody has a K D of 1.5 x 10 -8 or less as determined by surface plasmon resonance.
  • the present invention provides an anti-B7H3 antibody that binds to hB7-H3, said anti-B7H3 antibody comprising a heavy chain variable region comprising either a CDR set of SEQ ID NOs: 10, 11, and 12, and a light chain variable region comprising a CDR set of SEQ ID NOs: 14, 7, and 15; or a heavy chain variable region comprising a CDR set of SEQ ID NOs: 33, 35, and 35, and a light chain variable region comprising a CDR set of SEQ ID NOs: 37, 38, and 39.
  • the anti-B7H3 antibody is humanized.
  • the anti-B7H3 antibody further comprises a human acceptor framework.
  • the human acceptor framework comprises an amino acid sequence selected from the group consisting of SEQ ID Nos: 155, 156, 164, 165, 166, and 167. In one embodiment, the human acceptor framework comprises at least one framework region amino acid substitution. In one embodiment, the amino acid sequence of the framework is at least 65% identical to the sequence of the human acceptor framework and comprises at least 70 amino acid residues identical to the human acceptor framework. In one embodiment, the amino acid sequence of the framework is at least 85% identical, 90% identical, 95% identical, 96% identical, 97% identical, 98% identical, or 99% identical to the sequence of the human acceptor framework and comprises at least 70, at least 75, at least 80, or at least 85 amino acid residues identical to the human acceptor framework.
  • the present invention provides an anti-B7H3 antibody that binds to hB7-H3 comprising a heavy chain variable region comprising a CDR set of SEQ ID NOs: 25, 26, and 27, and a light chain variable region comprising a CDR set of SEQ ID NOs: 29, 30, and 31.
  • the anti-B7H3 antibody is humanized.
  • the antibody further comprises a human acceptor framework.
  • the human acceptor framework comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 155 to 158.
  • the present invention provides an anti-B7H3 antibody that binds to hB7-H3 comprising a heavy chain variable region comprising a CDR set of SEQ ID NOs: 33, 35, and 35, and a light chain variable region comprising a CDR set of SEQ ID NOs: 37, 38, and 39.
  • the anti-B7H3 antibod is humanized.
  • the anti-B7H3 antibody further comprises a human acceptor framework.
  • human acceptor framework comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 156, 158, 166 and 167.
  • the human acceptor framework comprises at least one framework region amino acid substitution at a key residue, said key residue selected from the group consisting of: a residue adjacent to a CDR; a glycosylation site residue; a rare residue; a residue capable of interacting with human B7-H3; a residue capable of interacting with a CDR; a canonical residue; a contact residue between heavy chain variable region and light chain variable region; a residue within a Vernier zone; and a residue in a region that overlaps between a Chothia-defined variable heavy chain CDR1 and a Kabat-defined first heavy chain framework.
  • the key residue is selected from the group consisting of 69H, 46L, 47L, 64L, and 71L (H refers to the heavy chain; L refers to the light chain; amino acid residues in reference to the Kabat numbering system).
  • the key residue substitution is in the variable heavy chain region and is L69I.
  • the key residue substitution is in the variable light chain region and is selected from the group consisting of L46P, L47W, G64V, and F71H.
  • the present invention provides an anti-hB7-H3 antibody comprising a heavy chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 10, a heavy chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 140, a heavy chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 12, a light chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 136 or 138, a light chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 7, and a light chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 15.
  • the present invention provides an anti-hB7-H3 antibody comprising a heavy chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 33, a heavy chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 34, a heavy chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 35, a light chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 37, a light chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 38, and a light chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 39.
  • the present invention provides an anti-hB7-H3 antibody comprising a heavy chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 139 and a light chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 135.
  • the present invention provides an anti-hB7-H3 antibody comprising a heavy chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 139, and/or a light chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 135.
  • the present invention provides an anti-hB7-H3 antibody comprising a heavy chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 139 and a light chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 137.
  • the present invention provides an anti-hB7-H3 antibody comprising a heavy chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 139, and/or a light chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 137.
  • the present invention provides an anti-hB7-H3 antibody comprising a heavy chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 147 and a light chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 144.
  • the present invention provides an anti-hB7-H3 antibody comprising a heavy chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 147, and/or a light chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 144.
  • the antibody, or antigen-binding portion thereof, provided herein binds to cynomolgus (cyno) B7-H3.
  • the antibody, or antigen binding portion thereof has a dissociation constant (K D ) to hB7-H3 selected from the group consisting of: at most about 10 -7 M; at most about 10 -8 M; at most about 10 -9 M; at most about 10 -10 M; at most about 10 -11 M; at most about 10 -12 M; and at most 10 -13 M.
  • K D dissociation constant
  • the antibody, or antigen binding portion thereof comprises a heavy chain immunoglobulin constant domain of a human IgM constant domain, a human IgG1 constant domain, a human IgG2 constant domain, a human IgG3 constant domain, a human IgG4 constant domain, a human IgA constant domain, or a human IgE constant domain.
  • the antibody is an IgG1 monoclonal antibody comprising a kappa light chain.
  • the human IgG1 constant domain comprises an amino acid sequence of SEQ ID NO: 159 or SEQ ID NO: 160.
  • the present invention provides an anti-hB7-H3 antibody comprising a sequence set selected from the group consisting of: a) a heavy chain comprising the amino acid sequence of SEQ ID NO: 168 and a light chain comprising the amino acid sequence of SEQ ID NO: 169; b) a heavy chain comprising the amino acid sequence of SEQ ID NO: 170 and a light chain comprising the amino acid sequence of SEQ ID NO: 171; and c) a heavy chain comprising the amino acid sequence of SEQ ID NO: 172 and a light chain comprising the amino acid sequence of SEQ ID NO: 173.
  • the anti-hB7-H3 antibody, or antigen-binding portion thereof comprises a heavy chain CDR set corresponding to antibody huAb13v1, and a light chain CDR set
  • the anti-hB7-H3 antibody, or antigen- binding portion thereof comprises a heavy chain variable region corresponding to antibody huAb13v1, and a light chain variable region corresponding to antibody huAb13v1.
  • the anti-hB7-H3 antibody, or antigen-binding portion thereof comprises a heavy chain CDR set corresponding to antibody huAb3v2.5, and a light chain CDR set
  • the anti-hB7-H3 antibody, or antigen- binding portion thereof comprises a heavy chain variable region corresponding to antibody huAb3v2.5, and a light chain variable region corresponding to antibody huAb3v2.5.
  • the anti-hB7-H3 antibody, or antigen-binding portion thereof competes with the antibody, or antigen binding portion thereof, of any one of the anti-hB7-H3 antibodies, or antigen-binding portions thereof, disclosed herein.
  • the anti-hB7-H3 antibody is an IgG, e.g., and IgG1, having four polypeptide chains which are two heavy chains and two light chains.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising an anti-hB7-H3 antibody, or antigen binding portion thereof, as disclosed herein, and a pharmaceutically acceptable carrier.
  • the present invention provides an anti-hB7-H3 Antibody Drug Conjugate (ADC) comprising an anti-hB7-H3 antibody disclosed herein conjugated to a drug via a linker.
  • the drug is an auristatin or a pyrrolobenzodiazepine (PBD).
  • the drug is a Bcl-xL inhibitor.
  • the present invention provides an anti-hB7-H3 antibody drug conjugate (ADC) comprising a drug linked to an anti-human B7-H3 (hB7-H3) antibody by way of a linker, wherein the drug is a Bcl-xL inhibitor according to structural formula (IIa) or (IIb):
  • ADC anti-hB7-H3 antibody drug conjugate
  • Z 1 is selected from N, CH, C-halo and C-CN;
  • Z 2a , Z 2b , and Z 2c are each, independent from one another, selected from a bond, NR 6 , CR 6a R 6b , O, S, S(O), SO 2 , NR 6 C(O), NR 6a C(O)NR 6b , and NR 6 C(O)O;
  • R 1 is selected from hydrogen, methyl, halo, halomethyl, ethyl and cyano;
  • R 2 is selected from hydrogen, methyl, halo, halomethyl and cyano;
  • R 3 is selected from hydrogen, lower alkyl and
  • R 6 , R 6a and R 6b are each, independent from one another, selected from hydrogen, lower alkyl, lower heteroalkyl, optionally substituted monocyclic cycloalklyl and monocyclic heterocyclyl, or are taken together with an atom from R 13 to form a cycloalkyl or heterocyclyl ring having between 3 and 7 ring atoms;
  • R 10 is selected from cyano, OR 14 , SR 14 , SOR 14 , SO 2 R 14 , SO 2 NR 14a R 14b , NR 14a R 14b , NHC(O)R 14 and NHSO 2 R 14 ;
  • R 11a and R 11b are each, independently of one another, selected from hydrogen, halo, methyl, ethyl, halomethyl, hydroxyl, methoxy, CN, and SCH 3 ;
  • R 12 is selected from cyano, OR 14 , SR 14 , SOR 14 , SO 2 R 14 , SO 2 NR 14
  • R 13 is selected from a bond, optionally substituted lower alkylene, optionally substituted lower heteroalkylene, optionally substituted cycloalkyl or optionally substituted heterocyclyl
  • R 14 is selected from hydrogen, optionally substituted lower alkyl and optionally substituted lower heteroalkyl
  • R 14a and R 14b are each, independently of one another, selected from hydrogen, optionally substituted lower alkyl, and optionally substituted lower heteroalkyl, or are taken together with the nitrogen atom to which they are bonded to form an optionally substituted monocyclic cycloalkyl or monocyclic heterocyclyl ring
  • R 15 is selected from hydrogen, halo, C 1-6 alkanyl, C 2-4 alkenyl, C 2-4 alkynyl, and C 1-4 haloalkyl and C 1-4 hydroxyalkyl, with the proviso that when R 15 is present, R 4 is not C 1-4 alkyl, C 2-4 alkenyl
  • the anti-hB7-H3 antibody either: comprises a heavy chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 10, a heavy chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 140, a heavy chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 12, a light chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 136 or 138, a light chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 7, and a light chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 15; or comprises a heavy chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 33, a heavy chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 34 a heavy chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO:
  • the ADC is a compound according to structural formula (I): wherein D is the Bcl-xL inhibitor drug of formula (IIa) or (IIb); L is the linker; Ab is the anti-hB7-H3 antibody; LK represents a covalent linkage linking the linker (L) to the anti-hB7-H3 antibody (Ab); and m is an integer ranging from 1 to 20.
  • the Ar 1 is unsubstituted. In one embodiment, the Ar 1 is
  • the Ar 2 is unsubstituted. In one embodiment, the Ar 2 is , which yl, C 1-4 alkoxy, and
  • Z 1 is N.
  • Z 2a is O.
  • R 1 is methyl or chloro.
  • R 2 is hydrogen or methyl. In one embodiment, R 2 is hydrogen.
  • R 4 is hydrogen or lower alkyl, wherein the lower alkyl is optionally substituted with C 1-4 alkoxy or C(O)NR 6a R 6b .
  • the drug is a Bcl-xL inhibitor according to structural formula (IIa). In one embodiment, the drug is a Bcl-xL inhibitor according to structural formula (IIa). In one embodiment, Z 2a is CH 2 or O. In one embodiment, R 13 is selected from lower alkylene or lower heteroalkylene.
  • the group .
  • Z 2a is oxygen
  • R 13 is CH 2 CH 2
  • R 4 is hydrogen or lower alkyl optionally substituted with C 1-4 alkoxy or C(O)NR 6a R 6b .
  • the ADC is a compound according to structural formula (IIb).
  • Z 2b is a bond, O, or NR 6 , or and R 13 is ethylene or optionally substituted heterocyclyl.
  • Z 2c is O and R 12 is lower alkyl optionally substituted with one or more halo or C 1-4 alkoxy.
  • the Bcl-xL inhibitor is selected from the group consisting of the following compounds modified in that the hydrogen corresponding to the # position of structural formula (IIa) or (IIb) is not present forming a monoradical: 6-[1-(1,3-benzothiazol-2-ylcarbamoyl)- 1,2,3,4-tetrahydroquinolin-7-yl]-3-[1-( ⁇ 3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.1 3,7 ]dec-1-yl ⁇ methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid; 6-[4-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-2H-1,4-benzoxazin-6-yl]-3-[1- ( ⁇ 3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.1 3,
  • the linker is cleavable by a lysosomal enzyme.
  • the lysosomal enzyme is Cathepsin B.
  • the linker comprises a segment according to structural formula (IVa), (IVb), (IVc), or (IVd):
  • peptide represents a peptide (illustrated N ⁇ C, wherein peptide includes the amino and carboxy“termini”) a cleavable by a lysosomal enzyme
  • T represents a polymer comprising one or more ethylene glycol units or an alkylene chain, or combinations thereof;
  • R a is selected from hydrogen, C 1-6 alkyl, SO 3 H and CH 2 SO 3 H;
  • R y is hydrogen or C 1-4 alkyl-(O) r -(C 1-4 alkylene) s -G 1 or C 1-4 alkyl-(N)-[(C 1-4 alkylene)-G 1 ] 2 ;
  • R z is C 1-4 alkyl-(O) r -(C 1-4 alkylene) s -G 2 ;
  • G 1 is SO 3 H, CO 2 H, PEG 4- 32, or sugar moiety;
  • G 2 is SO 3 H, CO 2 H, or PEG 4-32 moiety;
  • r
  • the peptide is selected from the group consisting of Val-Cit; Cit-Val; Ala-Ala; Ala-Cit; Cit-Ala; Asn-Cit; Cit-Asn; Cit-Cit; Val-Glu; Glu-Val; Ser-Cit; Cit-Ser; Lys-Cit; Cit-Lys; Asp-Cit; Cit-Asp; Ala-Val; Val-Ala; Phe-Lys; Lys-Phe; Val-Lys; Lys-Val; Ala-Lys; Lys- Ala; Phe-Cit; Cit-Phe; Leu-Cit; Cit-Leu; Ile-Cit; Cit-Ile; Phe-Arg; Arg-Phe; Cit-Trp; and Trp-Cit.
  • the lysosomal enzyme is ⁇ -glucuronidase or ⁇ -galactosidase.
  • the linker comprises a segment according to structural formula (Va), (Vb), (Vc), (Vd), or (Ve):
  • the linker comprises a segment according to structural formula (VIIIa), (VIIIb), or (VIIIc):
  • the linker comprises a polyethylene glycol segment having from 1 to 6 ethylene glycol units.
  • m is 2, 3 or 4.
  • the linker L comprises a segment according to structural formula (IVa) or (IVb).
  • the linker L is selected from the group consisting of IVa.1-IVa.8, IVb.1- IVb.19, IVc.1-IVc.7, IVd.1-IVd.4, Va.1-Va.12, Vb.1-Vb.10, Vc.1-Vc.11, Vd.1-Vd.6, Ve.1-Ve.2, VIa.1, VIc.1-V1c.2, VId.1-VId.4, VIIa.1-VIIa.4, VIIb.1-VIIb.8, VIIc.1-VIIc.6 in either the closed or open form.
  • the linker L is selected from the group consisting of IVb.2, IVc.5, IVc.6, IVc.7, IVd.4, Vb.9, Vc.11, VIIa.1, VIIa.3, VIIc.1, VIIc.4, and VIIc.5, wherein the maleimide of each linker has reacted with the antibody Ab, forming a covalent attachment as either a succinimide (closed form) or succinamide (open form).
  • the linker L is selected from the group consisting of IVb.2, IVc.5, IVc.6, IVd.4, Vc.11, VIIa.1, VIIa.3, VIIc.1, VIIc.4, VIIc.5, wherein the maleimide of each linker has reacted with the antibody Ab, forming a covalent attachment as either a succinimide (closed form) or succinamide (open form).
  • the linker L is selected from the group consisting of IVb.2, Vc.11, VIIa.3, IVc.6, and VIIc.1, wherein is the attachment point to drug D and @ is the attachment point to the LK, wherein when the linker is in the open form as shown below, @ can be either at the ⁇ -position or ⁇ -position of the carboxylic acid next to it:
  • LK is a linkage formed with an amino group on the anti-hB7-H3 antibody Ab.
  • LK is an amide or a thiourea.
  • LK is a linkage formed with a sulfhydryl group on the anti-hB7-H3 antibody Ab.
  • LK is a thioether
  • LK is selected from the group consisting of amide, thiourea and thioether; and m is an integer ranging from 1 to 8.
  • D is the Bcl-xL inhibitor as described herein (e.g., W3.01, W3.02, W3.03, W3.04, W3.05, W3.06, W3.07, W3.08, W3.09, W3.10, W3.11, W3.12, W3.13, W3.14, W3.15, W3.16, W3.17, W3.18, W3.19, W3.20, W3.21, W3.22, W3.23, W3.24, W3.25, W3.26, W3.27, W3.28, W3.29, W3.30, W3.31, W3.32, W3.33, W3.34, W3.35, W3.36, W3.37, W3.38, W3.39, W3.40, W3.41, W3.42, W3.43, and pharmaceutically acceptable salts thereof);
  • L is selected from the group consisting of linkers IVa.1-IVa.8, IVb.1-IVb.19, IVc.1-IVc.7, IVd.1-IVd.4, Va.1- Va.12, Vb.1-Vb.10, Vc.1-Vc.
  • D is the Bcl-xL inhibitor selected from the group consisting of the following compounds modified in that the hydrogen corresponding to the # position of structural formula (IIa) or (IIb) is not present, forming a monoradical: 3-(1- ⁇ [3-(2-aminoethoxy)-5,7- dimethyltricyclo[3.3.1.1 3,7 ]dec-1-yl]methyl ⁇ -5-methyl-1H-pyrazol-4-yl)-6-[1-(1,3-benzothiazol-2- ylcarbamoyl)-5,6-dihydroimidazo[1,5-a]pyrazin-7(8H)-yl]pyridine-2-carboxylic acid; 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-3-[1-( ⁇ 3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.1 3,7 ]de
  • the ADC is selected from the group consisting of huAb13v1-ZT, huAb13v1-ZZ, huAb13v1-SE, huAb13v1-SR, huAb3v2.5-ZT, huAb3v2.5-ZZ, huAb3v2.5-SE, huAb3v2.5-SR, huAb3v2.6-ZT, huAb3v2.6-ZZ, huAb3v2.6-SE, and huAb3v2.6-SR, wherein huAb13v1, huAb3v2.5, and huAb3v2.6 are the anti-hB7-H3 antibodies and KZ, SR, SE, XW, YG, ZT and ZZ are synthons disclosed in Table B, and wherein the conjugated synthons are either in open or closed form.
  • Ab is an anti-hB7-H3 antibody, wherein the anti-hB7-H3 antibody comprises a heavy chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 35, a heavy chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 34, and a heavy chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 33; and a light chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 39, a light chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 38, and a light chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 37.
  • the Ab is an anti-hB7-H3 antibody, wherein the anti-hB7H3 antibody comprises a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 147, and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 144.
  • Ab is an anti-hB7-H3 antibody, wherein the anti-hB7-H3 antibody comprises a heavy chain constant region comprising the amino acid sequence set forth in SEQ ID NO: 160 and/or a light chain constant region comprising the amino acid sequence set forth in SEQ ID NO: 161.
  • Ab is an anti-hB7-H3 antibody, wherein the anti-hB7-H3 antibody comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 168, and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 169.
  • Ab is an anti-hB7-H3 antibody, wherein the anti-hB7-H3 antibody comprises a heavy chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 12, a heavy chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 140, and a heavy chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 10; and a light chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 15, a light chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 7, and a light chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 136.
  • the anti-hB7-H3 antibody comprises a heavy chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 12, a heavy chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 140, and a heavy chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO:
  • the Ab is an anti-hB7-H3 antibody, wherein the anti-hB7H3 antibody comprises a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 139, and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 135.
  • Ab is an anti-hB7-H3 antibody, wherein the anti-hB7-H3 antibody comprises a heavy chain constant region comprising the amino acid sequence set forth in SEQ ID NO: 160 and/or a light chain constant region comprising the amino acid sequence set forth in SEQ ID NO: 161.
  • Ab is an anti-hB7-H3 antibody, wherein the anti-hB7-H3 antibody comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 170, and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 171.
  • n is an integer from 2 to 6. In one embodiment, m is 2.
  • the ADC comprises an anti-hB7-H3 antibody comprising a heavy chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 12, a heavy chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 140, and a heavy chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 10; a light chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 15, a light chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 7, and a light chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 136 or 138.
  • the ADC comprises an antibody comprising a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 139, and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 135.
  • the ADC comprises an antibody comprising a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 139, and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 137.
  • the ADC comprises an antibody comprising a light chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 39, a light chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 38, and a light chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 37; and a heavy chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 35 a heavy chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 34, and a heavy chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 33.
  • the ADC comprises an antibody comprising a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 147, and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 144.
  • the ADC is selected from the group consisting of huAb3v2.5-ZT, huAb3v2.5-ZZ, huAb3v2.5-XW, huAb3v2.5-SE, huAb3v2.5-SR, huAb3v2.5-YG, huAb3v2.5-KZ, huAb3v2.6-ZT, huAb3v2.6-ZZ, huAb3v2.6-XW, huAb3v2.6-SE, huAb3v2.6-SR, huAb3v2.6-YG, huAb3v2.6-KZ, huAb13v1-ZT, huAb13v1-ZZ, huAb13v1-XW, huAb13v1-SE, huAb13v1-SR, huAb13v1-YG, and huAb13v1-KZ.
  • the present invention provides a pharmaceutical composition comprising an effective amount of an ADC described herein, and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition comprises an ADC mixture comprising a plurality of the ADCs described herein, and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition comprises an ADC mixture having an average drug to antibody ratio (DAR) of 1.5 to 4.
  • DAR drug to antibody ratio
  • the pharmaceutical composition comprises an ADC mixture comprising ADCs each having a DAR of 1.5 to 8.
  • the present invention provides a method for treating cancer, comprising administering a therapeutically effective amount of the ADC described herein to a subject in need thereof.
  • the cancer is selected from the group consisting of small cell lung cancer, non small cell lung cancer, breast cancer, ovarian cancer, a glioblastoma, prostate cancer, pancreatic cancer, colon cancer, gastric cancer, melanoma, hepatocellular carcinoma, head and neck cancer, kidney cancer, leukemia, e.g., acute myeloid leukemia (AML), and lymphoma, e.g., non-Hodgkin's lymphoma (NHL).
  • the cancer is a squamous cell carcinoma. In one embodiment
  • the squamous cell carcinoma is squamous lung cancer or squamous head and neck cancer.
  • the cancer is triple negative breast cancer.
  • the cancer is non-small cell lung cancer.
  • the present invention provides a method for inhibiting or decreasing solid tumor growth in a subject having a solid tumor, said method comprising administering an effective amount of the ADC described herein to the subject having the solid tumor, such that the solid tumor growth is inhibited or decreased.
  • the solid tumor is a non-small cell lung carcinoma.
  • the cancer is characterized as having an activating EGFR mutation.
  • the activating EGFR mutation is selected from the group consisting of an exon 19 deletion mutation, a single-point substitution mutation L858R in exon 21, a T790M point mutation, and combinations thereof.
  • the ADC is administered in combination with an additional agent or an additional therapy.
  • the additional agent is selected from the group consisting of an anti-PD1 antibody (e.g. pembrolizumab), an anti-PD-L1 antibody (e.g. atezolizumab), an anti- CTLA-4 antibody (e.g. ipilimumab), a MEK inhibitor (e.g. trametinib), an ERK inhibitor, a BRAF inhibitor (e.g. dabrafenib), osimertinib, erlotinib, gefitinib, sorafenib, a CDK9 inhibitor (e.g.
  • a MCL-1 inhibitor temozolomide
  • a Bcl-xL inhibitor e.g. venetoclax
  • ibrutinib e.g. everolimus
  • a mTOR inhibitor e.g. everolimus
  • a PI3K inhibitor e.g. buparlisib
  • duvelisib idelalisib
  • an AKT inhibitor e.g. HER2 inhibitor (e.g. lapatinib), a taxane (e.g. docetaxel, paclitaxel, nab- paclitaxel), an ADC comprising an auristatin, an ADC comprising a PBD (e.g.
  • an ADC comprising a maytansinoid (e.g. TDM1), a TRAIL agonist, a proteasome inhibitor (e.g. bortezomib), and a nicotinamide phosphoribosyltransferase (NAMPT) inhibitor.
  • a maytansinoid e.g. TDM1
  • a TRAIL agonist e.g. a TRAIL agonist
  • a proteasome inhibitor e.g. bortezomib
  • NAMPT nicotinamide phosphoribosyltransferase
  • the additional therapy is radiation.
  • the additional agent is a chemotherapeutic agent.
  • the anti-B7-H3 ADCs of the invention are administered in combination with venetoclax to a human subject for the treatment of small cell lung cancer (SCLC).
  • SCLC small cell lung cancer
  • the present invention provides a process for the preparation of an ADC according to structural formula (I): wherein:
  • D is the Bcl-xL inhibitor drug of formula (IIa) or (IIb) as disclosed herein;
  • L is the linker as disclosed herein;
  • Ab is an hB7-H3 antibody, wherein the hB7-H3 antibody comprises the heavy and light chain CDRs of huAb3v2.5, huAb3v2.6, or huAb13v1;
  • LK represents a covalent linkage linking linker L to antibody Ab
  • n is an integer ranging from 1 to 20;
  • ADC is optionally purified by hydrophobic interaction chromatography.
  • n is 2.
  • the present invention provides an ADC prepared by the process as described above.
  • Figure 1 is a graphical representation of the epitope grouping of murine anti-B7-H3 hybridoma antibodies as determined by pair-wise binding assays.
  • Figure 2 depicts an antibody reduction, modification with a maleimide derivative to give a thiosuccinimide intermediate, and subsequent hydrolysis of thiosuccinimide moiety
  • Figure 3 depicts the structure of an antibody-maleimidocaproyl-vc-PABA-MMAE ADC.
  • Figure 4 depicts the structure of a PBD dimer (SGD-1882) conjugated to an antibody (Ab) via a maleimidocaproyl-valine-alanine linker (collectively referred to as SGD-1910).
  • Figure 5 depicts the MS characterization of light chain and heavy chain of huAb13v11) prior to conjugation, 2) after conjugation to a maleimide derivative to give a thiosuccinimide intermediate and 3) post pH 8-mediated hydrolysis of the thiosuccinimide ring.
  • Various aspects of the invention relate to anti-B7-H3 antibodies and antibody fragments, anti- B7-H3 ADCs, and pharmaceutical compositions thereof, as well as nucleic acids, recombinant expression vectors and host cells for making such antibodies and fragments.
  • Methods of using the antibodies, fragments, and ADCs described herein to detect human B7-H3, to inhibit human B7-H3 activity (in vitro or in vivo), and to treat cancers are also encompassed by the invention.
  • the invention provides anti-B7-H3 ADCs, including ADCs comprising Bcl-xL inhibitors, synthons useful for synthesizing the ADCs, compositions comprising the ADCs, methods of making the ADCs, and various methods of using the ADCs.
  • the ADCs disclosed herein are“modular” in nature.
  • various specific embodiments of the various“modules” comprising the ADCs, as well as the synthons useful for synthesizing the ADCs are described.
  • specific embodiments of antibodies, linkers, and Bcl-xL inhibitors that may comprise the ADCs and synthons are described. It is intended that all of the specific embodiments described may be combined with each other as though each specific combination were explicitly described individually.
  • ADCs and/or ADC synthons described herein may be in the form of salts, and in certain embodiments, particularly pharmaceutically acceptable salts.
  • the compounds of the present disclosure that possess a sufficiently acidic, a sufficiently basic, or both functional groups can react with any of a number of inorganic bases, and inorganic and organic acids, to form a salt.
  • compounds that are inherently charged, such as those with a quaternary nitrogen can form a salt with an appropriate counterion, e.g., a halide such as a bromide, chloride, or fluoride.
  • Acids commonly employed to form acid addition salts are inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, phosphoric acid, and the like, and organic acids such as p-toluenesulfonic acid, methanesulfonic acid, oxalic acid, p-bromophenyl- sulfonic acid, carbonic acid, succinic acid, citric acid, etc.
  • Base addition salts include those derived from inorganic bases, such as ammonium and alkali or alkaline earth metal hydroxides, carbonates, bicarbonates, and the like.
  • anti-B7-H3 antibody refers to an antibody that specifically binds to B7-H3.
  • An antibody“which binds” an antigen of interest, i.e., B7-H3, is one capable of binding that antigen with sufficient affinity such that the antibody is useful in targeting a cell expressing the antigen.
  • the antibody specifically binds to human B7-H3 (hB7-H3). Examples of anti- B7-H3 antibodies are disclosed in the examples below.
  • anti- B7-H3 antibody refers to an antibody which binds to wild type B7-H3 (e.g., a 4IgB7-H3 isoform of B7-H3) or any variant of B7-H3.
  • wild type B7-H3 e.g., a 4IgB7-H3 isoform of B7-H3
  • amino acid sequence of wild type human B7-H3 is provided below as SEQ ID NO: 149, where the signal peptide (amino acid residues 1-28) is underlined.
  • the extracellular domain (ECD) of human B7-H3 is provided in SEQ ID NO: 152 (inclusive of a His tag).
  • the antibody of ADC binds the ECD of human B7-H3 as described in the ECD of SEQ ID NO: 152.
  • a particular structure e.g., an antigenic determinant or epitope
  • an antibody "binds specifically" to a target if the antibody, when labeled, can be competed away from its target by the corresponding non-labeled antibody.
  • an antibody specifically binds to a target, e.g., B7-H3, if the antibody has a K D for the target of at least about 10 -4 M, 10 -5 M, 10 -6 M, 10-7 M, 10 -8 M, 10 -9 M, 10 -10 M, 10 -11 M, 10 -12 M, or less (less meaning a number that is less than 10- 12 , e.g.10 -13 ).
  • the term“specific binding to B7-H3” or“specifically binds to B7- H3,” as used herein, refers to an antibody or an ADC that binds to B7-H3 and has a dissociation constant (K D ) of 1.0 x 10 -7 M or less, as determined by surface plasmon resonance. It shall be understood, however, that the antibody or ADC may be capable of specifically binding to two or more antigens which are related in sequence. For example, in one embodiment, an antibody can specifically bind to both human and a non-human (e.g., mouse or non-human primate) orthologs of B7-H3.
  • K D dissociation constant
  • antibody refers to an immunoglobulin molecule that specifically binds to an antigen and comprises a heavy (H) chain(s) and a light (L chain(s).
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, CH1, CH2 and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region.
  • the light chain constant region is comprised of one domain, CL.
  • VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • Each VH and VL is composed of three CDRs and four FRs, arranged from amino- terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • An antibody can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY) and class (e.g., IgG1, IgG2, IgG 3, IgG4, IgA1 and IgA2) or subclass.
  • an antibody is not intended to include antigen binding portions of an antibody (defined below), it is intended, in certain embodiments, to describe an antibody comprising a small number of amino acid deletions from the carboxy end of the heavy chain(s).
  • an antibody comprises a heavy chain having 1-5 amino acid deletions the carboxy end of the heavy chain.
  • an antibody is a monoclonal antibody which is an IgG, having four polypeptide chains, two heavy (H) chains, and two light (L chains) that can bind to hB7-H3.
  • an antibody is a monoclonal IgG antibody comprising a lambda or a kappa light chain.
  • antibody portion or“antigen binding fragment” of an antibody (or simply “antibody portion” or“antibody fragment”), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., hB7-H3). It has been shown that the antigen binding function of an antibody can be performed by fragments of a full-length antibody. Such antibody embodiments may also be bispecific, dual specific, or multi-specific formats; specifically binding to two or more different antigens.
  • binding fragments encompassed within the term“antigen binding portion” of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab') 2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341:544- 546, Winter et al., PCT publication WO 90/05144 A1 herein incorporated by reference), which comprises a single variable domain; and (vi) an isolated complementarity determining region (CDR).
  • CDR complementarity determining region
  • the two domains of the Fv fragment, VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883).
  • single chain Fv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term“antigen binding portion” of an antibody.
  • scFv molecules may be incorporated into a fusion protein.
  • Other forms of single chain antibodies, such as diabodies are also encompassed.
  • Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R.J., et al. (1994) Structure 2:1121-1123).
  • Such antibody binding portions are known in the art (Kontermann and Dubel eds., Antibody Engineering (2001) Springer-Verlag. New York.790 pp. (ISBN 3-540-41354-5).
  • IgG immunoglobuin G
  • IgG heavy chain and light chain constant domain amino acid sequences are known in the art and represented below in Table A. Table A: Sequences of human IgG heavy chain constant domains and light chain constant domains
  • an“isolated antibody”, as used herein, is intended to refer to an antibody that is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds B7-H3 is substantially free of antibodies that specifically bind antigens other than B7-H3).
  • An isolated antibody that specifically binds B7-H3 may, however, have cross-reactivity to other antigens, such as B7-H3 molecules from other species.
  • an isolated antibody may be substantially free of other cellular material and/or chemicals.
  • humanized antibody refers to antibodies which comprise heavy and light chain variable region sequences from a nonhuman species (e.g., a mouse) but in which at least a portion of the VH and/or VL sequence has been altered to be more“human-like”, i.e., more similar to human germline variable sequences.
  • the term“humanized antibody” is an antibody or a variant, derivative, analog or fragment thereof which immunospecifically binds to an antigen of interest and which comprises a framework (FR) region having substantially the amino acid sequence of a human antibody and a complementary determining region (CDR) having substantially the amino acid sequence of a non-human antibody.
  • FR framework
  • CDR complementary determining region
  • the term“substantially” in the context of a CDR refers to a CDR having an amino acid sequence at least 80%, preferably at least 85%, at least 90%, at least 95%, at least 98% or at least 99% identical to the amino acid sequence of a non-human antibody CDR.
  • a humanized antibody comprises substantially all of at least one, and typically two, variable domains (Fab, Fab', F(ab') 2 , FabC, Fv) in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin (i.e., donor antibody) and all or substantially all of the framework regions are those of a human immunoglobulin consensus sequence.
  • a humanized antibody also comprises at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • a humanized antibody contains both the light chain as well as at least the variable domain of a heavy chain.
  • the antibody also may include the CH1, hinge, CH2, CH3, and CH4 regions of the heavy chain.
  • a humanized antibody only contains a humanized light chain.
  • a humanized antibody only contains a humanized heavy chain.
  • a humanized antibody only contains a humanized variable domain of a light chain and/or humanized heavy chain.
  • the humanized antibody can be selected from any class of immunoglobulins, including IgM, IgG IgD IgA and IgE and any isotype including without limitation IgG1 IgG2 IgG3 and IgG4
  • the humanized antibody may comprise sequences from more than one class or isotype, and particular constant domains may be selected to optimize desired effector functions using techniques well-known in the art.
  • Kabat numbering “Kabat definitions,” and“Kabat labeling” are used interchangeably herein. These terms, which are recognized in the art, refer to a system of numbering amino acid residues which are more variable (i.e., hypervariable) than other amino acid residues in the heavy and light chain variable regions of an antibody, or an antigen binding portion thereof (Kabat et al. (1971) Ann. NY Acad, Sci.190:382-391 and, Kabat, E.A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH
  • the hypervariable region ranges from amino acid positions 31 to 35 for CDR1, amino acid positions 50 to 65 for CDR2, and amino acid positions 95 to 102 for CDR3.
  • the hypervariable region ranges from amino acid positions 24 to 34 for CDR1, amino acid positions 50 to 56 for CDR2, and amino acid positions 89 to 97 for CDR3.
  • CDR refers to the complementarity determining region within antibody variable sequences. There are three CDRs in each of the variable regions of the heavy chain (HC) and the light chain (LC), which are designated CDR1, CDR2 and CDR3 (or specifically HC CDR1, HC CDR2, HC CDR3, LC CDR1, LC CDR2, and LC CDR3), for each of the variable regions.
  • CDR set refers to a group of three CDRs that occur in a single variable region capable of binding the antigen. The exact boundaries of these CDRs have been defined differently according to different systems.
  • CDR boundary definitions may not strictly follow one of the above systems, but will nonetheless overlap with the Kabat CDRs, although they may be shortened or lengthened in light of prediction or experimental findings that particular residues or groups of residues or even entire CDRs do not significantly impact antigen binding.
  • the methods used herein may utilize CDRs defined according to any of these systems although preferred embodiments use Kabat or Chothia defined CDRs
  • the term“framework” or“framework sequence” refers to the remaining sequences of a variable region minus the CDRs. Because the exact definition of a CDR sequence can be determined by different systems, the meaning of a framework sequence is subject to
  • the six CDRs (CDR-L1, CDR-L2, and CDR-L3 of light chain and CDR-H1, CDR-H2, and CDR-H3 of heavy chain) also divide the framework regions on the light chain and the heavy chain into four sub-regions (FR1, FR2, FR3 and FR4) on each chain, in which CDR1 is positioned between FR1 and FR2, CDR2 between FR2 and FR3, and CDR3 between FR3 and FR4.
  • a framework region represents the combined FR's within the variable region of a single, naturally occurring immunoglobulin chain.
  • a FR represents one of the four sub- regions
  • FRs represents two or more of the four sub- regions constituting a framework region.
  • the framework and CDR regions of a humanized antibody need not correspond precisely to the parental sequences, e.g., the donor antibody CDR or the consensus framework may be mutagenized by substitution, insertion and/or deletion of at least one amino acid residue so that the CDR or framework residue at that site does not correspond to either the donor antibody or the consensus framework. In a preferred embodiment, such mutations, however, will not be extensive. Usually, at least 80%, preferably at least 85%, more preferably at least 90%, and most preferably at least 95% of the humanized antibody residues will correspond to those of the parental FR and CDR sequences.
  • the term“consensus framework” refers to the framework region in the consensus immunoglobulin sequence.
  • the term“consensus immunoglobulin sequence” refers to the sequence formed from the most frequently occurring amino acids (or nucleotides) in a family of related immunoglobulin sequences (See e.g., Winnaker, From Genes to Clones (Verlagsgesellschaft, Weinheim, Germany 1987). In a family of immunoglobulins, each position in the consensus sequence is occupied by the amino acid occurring most frequently at that position in the family. If two amino acids occur equally frequently, either can be included in the consensus sequence.
  • human acceptor framework is meant to refer to a framework of an antibody or antibody fragment thereof comprising the amino acid sequence of a VH or VL framework derived from a human antibody or antibody fragment thereof or a human consensus sequence framework into which CDRs from a non-human species may be incorporated.
  • Percent (%) amino acid sequence identity with respect to a peptide or polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the specific peptide or polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art for instance using publicly available computer software such as BLAST BLAST-2 ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
  • the invention includes an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identity to an amino acid sequence set forth in any one of SEQ ID NOs: 1 to 148.
  • multivalent antibody is used herein to denote an antibody comprising two or more antigen binding sites.
  • the multivalent antibody may be engineered to have the three or more antigen binding sites, and is generally not a naturally occurring antibody.
  • multispecific antibody refers to an antibody capable of binding two or more unrelated antigens.
  • the multispecific antibody is a bispecific antibody that is capable of binding to two unrelated antigens, e.g., a bispecific antibody, or antigen-binding portion thereof, that binds B7-H3 and CD3.
  • DVD dual variable domain
  • Such DVDs may be monospecific, i.e., capable of binding one antigen or multispecific, i.e. capable of binding two or more antigens.
  • DVD binding proteins comprising two heavy chain DVD polypeptides and two light chain DVD polypeptides are referred to a DVD Ig. Each half of a DVD Ig comprises a heavy chain DVD polypeptide, and a light chain DVD
  • Each binding site comprises a heavy chain variable domain and a light chain variable domain with a total of 6 CDRs involved in antigen binding per antigen binding site.
  • the CDRs described herein are used in an anti-B7-H3 DVD.
  • chimeric antigen receptor refers to a recombinant protein comprising at least (1) an antigen-binding region, e.g., a variable heavy or light chain of an antibody, (2) a transmembrane domain to anchor the CAR into a T cell, and (3) one or more intracellular signaling domains.
  • activity includes activities such as the binding specificity/affinity of an antibody or ADC for an antigen, for example, an anti-hB7-H3 antibody that binds to an hB7-H3 antigen and/or the neutralizing potency of an antibody, for example, an anti-hB7-H3 antibody whose binding to hB7- H3 inhibits the biological activity of hB7-H3, e.g., inhibition of proliferation of B7-H3 expressing cell lines, e.g., human H146 lung carcinoma cells, human H1650 lung carcinoma cells, or human EBC1 lung carcinoma cells.
  • B7-H3 expressing cell lines e.g., human H146 lung carcinoma cells, human H1650 lung carcinoma cells, or human EBC1 lung carcinoma cells.
  • non small-cell lung carcinoma (NSCLC) xenograft assay refers to an in vivo assay used to determine whether an anti-B7-H3 antibody or ADC, can inhibit tumor growth (e.g., further growth) and/or decrease tumor growth resulting from the transplantation of NSCLC cells into an immunodeficient mouse.
  • An NSCLC xenograft assay includes transplantation of NSCLC cells into an immunodeficient mouse such that a tumor grows to a desired size e g 200-250 mm 3 whereupon the antibody or ADC is administered to the mouse to determine whether the antibody or ADC can inhibit and/or decrease tumor growth.
  • the activity of the antibody or ADC is determined according to the percent tumor growth inhibition (%TGI) relative to a control antibody, e.g., a human IgG antibody (or collection thereof) which does not specifically bind tumor cells, e.g., is directed to an antigen not associated with cancer or is obtained from a source which is noncancerous (e.g., normal human serum).
  • a control antibody e.g., a human IgG antibody (or collection thereof) which does not specifically bind tumor cells, e.g., is directed to an antigen not associated with cancer or is obtained from a source which is noncancerous (e.g., normal human serum).
  • the antibody (or ADC) and the control antibody are administered to the mouse at the same dose, with the same frequency, and via the same route.
  • the mouse used in the NSCLC xenograft assay is a severe combined immunodeficiency (SCID) mouse and/or an athymic CD-1 nude mouse.
  • SCID severe combined
  • H1299 cells NCI- H1299 [H-1299] (ATCC ® CRL-5803)
  • H1650 cells NCI-H1650 [H-1650] (ATCC ® CRL-5883TM)
  • H1975 cells NCI-H1975 cells [H1975] (ATCC ® CRL-5908TM)
  • EBC-1 cells EBC-1 cells.
  • SCLC xenograft assay refers to an in vivo assay used to determine whether an anti-B7-H3 antibody or ADC, can inhibit tumor growth (e.g., further growth) and/or decrease tumor growth resulting from the transplantation of SCLC cells into an immunodeficient mouse.
  • An SCLC xenograft assay includes transplantation of SCLC cells into an immunodeficient mouse such that a tumor grows to a desired size, e.g., 200-250 mm 3 , whereupon the antibody or ADC is administered to the mouse to determine whether the antibody or ADC can inhibit and/or decrease tumor growth.
  • the activity of the antibody or ADC is determined according to the percent tumor growth inhibition (%TGI) relative to a control antibody, e.g., a human IgG antibody (or collection thereof) which does not specifically bind tumor cells, e.g., is directed to an antigen not associated with cancer or is obtained from a source which is noncancerous (e.g., normal human serum).
  • a control antibody e.g., a human IgG antibody (or collection thereof) which does not specifically bind tumor cells, e.g., is directed to an antigen not associated with cancer or is obtained from a source which is noncancerous (e.g., normal human serum).
  • the antibody (or ADC) and the control antibody are administered to the mouse at the same dose, with the same frequency, and via the same route.
  • the mouse used in the NSCLC xenograft assay is a severe combined
  • SCLC cells immunodeficiency (SCID) mouse and/or an athymic CD-1 nude mouse.
  • SCLC cells that may be used in the SCLC xenograft assay include, but are not limited to, H146 cells (NCI-H146 cells [H146] (ATCC ® HTB-173TM)), and H847 cells (NCI-H847 [H847] (ATCC ® CRL-5846TM)).
  • epitope refers to a region of an antigen that is bound by an antibody or ADC.
  • epitope determinants include chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl, or sulfonyl, and, in certain embodiments, may have specific three dimensional structural characteristics, and/or specific charge characteristics.
  • an antibody is said to specifically bind an antigen when it preferentially recognizes its target antigen in a complex mixture of proteins and/or macromolecules.
  • surface plasmon resonance refers to an optical phenomenon that allows for the analysis of real-time biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix for example using the BIAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, NJ).
  • BIAcore Pharmaacia Biosensor AB, Uppsala, Sweden and Piscataway, NJ.
  • surface plasmon resonance is determined according to the methods described in Example 2.
  • k off or“ k d ”, as used herein, is intended to refer to the off rate constant for dissociation of an antibody from the antibody/antigen complex.
  • K D is intended to refer to the equilibrium dissociation constant of a particular antibody-antigen interaction (e.g., huAb13 antibody and B7-H3). K D is calculated by k a / k d .
  • competitive binding refers to a situation in which a first antibody competes with a second antibody, for a binding site on a third molecule, e.g., an antigen.
  • competitive binding between two antibodies is determined using FACS analysis.
  • a competitive binding assay is an assay used to determine whether two or more antibodies bind to the same epitope.
  • a competitive binding assay is a competition fluorescent activated cell sorting (FACS) assay which is used to determine whether two or more antibodies bind to the same epitope by determining whether the fluorescent signal of a labeled antibody is reduced due to the introduction of a non-labeled antibody, where competition for the same epitope will lower the level of fluorescence.
  • FACS competition fluorescent activated cell sorting
  • label refers to an antibody, or an antigen binding portion thereof, with a label incorporated that provides for the identification of the binding protein, e.g., an antibody.
  • the label is a detectable marker, e.g., incorporation of a radiolabeled amino acid or attachment to a polypeptide of biotinyl moieties that can be detected by marked avidin (e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or colorimetric methods).
  • markers for polypeptides include, but are not limited to, the following: radioisotopes or radionuclides (e.g., 3 H 14
  • fluorescent labels e.g., FITC, rhodamine, lanthanide phosphors
  • enzymatic labels e.g., horseradish peroxidase, luciferase, alkaline phosphatase
  • chemiluminescent markers e.g., biotinyl groups
  • predetermined polypeptide epitopes recognized by a secondary reporter e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags
  • magnetic agents such as gadolinium chelates.
  • an ADC refers to a binding protein, such as an antibody or antigen binding fragment thereof, chemically linked to one or more chemical drug(s) (also referred to herein as agent(s), warhead(s), or payload(s)) that may optionally be therapeutic or cytotoxic agents
  • an ADC includes an antibody a drug (e g a cytotoxic drug), and a linker that enables attachment or conjugation of the drug to the antibody.
  • An ADC typically has anywhere from 1 to 8 drugs conjugated to the antibody, including drug loaded species of 2, 4, 6, or 8.
  • Non-limiting examples of drugs that may be included in the ADCs are mitotic inhibitors, antitumor antibiotics, immunomodulating agents, vectors for gene therapy, alkylating agents, antiangiogenic agents, antimetabolites, boron-containing agents, chemoprotective agents, hormones, antihormone agents, corticosteroids, photoactive therapeutic agents, oligonucleotides, radionuclide agents, topoisomerase inhibitors, kinase inhibitors (e.g., TEC-family kinase inhibitors and serine/threonine kinase inhibitors), and radiosensitizers.
  • the drug is a Bcl-xL inhibitor.
  • the anti- B7-H3 ADC comprises antibody huAb13v1, huAb3v2.5, or huAb3v2.6 conjugated to an auristatin, e.g., MMAE or MMAF.
  • the anti-B7-H3 ADC comprises antibody huAb13v1, huAb3v2.5, or huAb3v2.6 conjugated to a Bcl-xL inhibitor.
  • the anti-B7- H3B7-H3 ADC binds to human B7-H3B7-H3B7-H3.
  • Bcl-xL inhibitor refers to a compound which antagonizes Bcl-xL activity in a cell.
  • a Bcl-xL inhibitor promotes apoptosis of a cell by inhibiting Bcl-xL activity.
  • auristatin refers to a family of antimitotic agents. Auristatin derivatives are also included within the definition of the term“auristatin”. Examples of auristatins include, but are not limited to, auristatin E (AE), monomethylauristatin E (MMAE),
  • MMAF monomethylauristatin F
  • an anti- B7-H3 antibody described herein is conjugated to an auristatin to form an anti-B7-H3 ADC.
  • the term“Ab-vcMMAE” is used to refer to an ADC comprising an antibody conjugated to monomethylauristatin E (MMAE) via a maleimidocaproyl valine citrulline p- aminobenzyloxycarbamyl (PABA) linker.
  • MMAE monomethylauristatin E
  • PABA p- aminobenzyloxycarbamyl
  • mcMMAF is used to refer to a linker/drug combination of maleimidocaproyl-monomethylauristatin F (MMAF).
  • DAR drug-to-antibody ratio
  • the term“drug-to-antibody ratio” or“DAR” refers to the number of drugs, e.g., a Bcl-xL inhibitor, attached to the antibody of the ADC.
  • the DAR of an ADC can range from 1 to 8, although higher loads, e.g., 20, are also possible depending on the number of linkage site on an antibody.
  • the term DAR may be used in reference to the number of drugs loaded onto an individual antibody, or, alternatively, may be used in reference to the average or mean DAR of a group of ADCs.
  • undesired ADC species refers to any drug loaded species which is to be separated from an ADC species having a different drug load.
  • the term undesired ADC species may refer to drug loaded species of 6 or more i e ADCs with a DAR of 6 or more, including DAR6, DAR7, DAR8, and DAR greater than 8 (i.e., drug loaded species of 6, 7, 8, or greater than 8).
  • the term undesired ADC species may refer to drug loaded species of 8 or more, i.e., ADCs with a DAR of 8 or more, including DAR8, and DAR greater than 8 (i.e., drug loaded species of 8, or greater than 8).
  • ADC mixture refers to a composition containing a heterogeneous DAR distribution of ADCs.
  • an ADC mixture contains ADCs having a distribution of DARs of 1 to 8, e.g., 1.5, 2, 4, 6, and 8 (i.e., drug loaded species of 1.5, 2, 4, 6, and 8).
  • DARs 1 to 8
  • degradation products may result such that DARs of 1, 3, 5, and 7 may also be included in the mixture.
  • ADCs within the mixture may also have DARs greater than 8.
  • the ADC mixture results from interchain disulfide reduction followed by conjugation.
  • the ADC mixture comprises both ADCs with a DAR of 4 or less (i.e., a drug loaded species of 4 or less) and ADCs with a DAR of 6 or more (i.e., a drug loaded species of 6 or more).
  • a“xenograft assay” refers to a human tumor xenograft assay, wherein human tumor cells are transplanted, either under the skin or into the organ type in which the tumor originated, into immunocompromised mice that do not reject human cells.
  • cancer is meant to refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies.
  • cancers include glioblastoma, acute myeloid leukemia (AML), non- Hodgkin's lymphoma (NHL), non-small cell lung cancer, lung cancer, colon cancer, colorectal cancer, head and neck cancer, breast cancer (e.g., triple negative breast cancer), pancreatic cancer, squamous cell tumors, squamous cell carcinoma (e.g., squamous cell lung cancer or squamous cell head and neck cancer), anal cancer, skin cancer, and vulvar cancer.
  • the antibodies or ADCs of the invention are administered to a patient having a tumor(s) that overexpresses B7-H3.
  • the antibodies or ADCs of the invention are administered to a patient having a solid tumor which is likely to overexpress B7-H3. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having squamous cell non-small cell lung cancer (NSCLC). In one embodiment, the antibodies or ADCs of the invention are administered to a patient having solid tumors, including advanced solid tumors. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having prostate cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having non-small cell lung cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having a glioblastoma.
  • NSCLC squamous cell non-small cell lung cancer
  • the antibodies or ADCs of the invention are administered to a patient having colon cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having head and neck cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having kidney cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having clear cell renal cell carcinoma. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having glioma. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having melanoma. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having pancreatic cancer.
  • the antibodies or ADCs of the invention are administered to a patient having gastric cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having ovarian cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having colorectal cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having renal cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having small cell lung cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having hepatocellular carcinoma. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having hypopharyngeal squamous cell carcinoma.
  • the antibodies or ADCs of the invention are administered to a patient having neuroblastoma. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having breast cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having endometrial cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having urothelial cell carcinoma. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having acute myeloid leukemia (AML). In one embodiment, the antibodies or ADCs of the invention are administered to a patient having non-Hodgkin's lymphoma (NHL).
  • AML acute myeloid leukemia
  • B7-H3 expressing tumor refers to a tumor which expresses B7-H3 protein.
  • B7-H3 expression in a tumor is determined using immunohistochemical staining of tumor cell membranes, where any immunohistochemical staining above background level in a tumor sample indicates that the tumor is a B7-H3 expressing tumor.
  • Methods for detecting expression of B7-H3 in a tumor are known in the art, and include immunohistochemical assays.
  • a“B7-H3 negative tumor” is defined as a tumor having an absence of B7-H3 membrane staining above background in a tumor sample as determined by immunohistochemical techniques.
  • overexpress “overexpression,” or“overexpressed” interchangeably refer to a gene that is transcribed or translated at a detectably greater level, usually in a cancer cell, in comparison to a normal cell.
  • Overexpression therefore refers to both overexpression of protein and RNA (due to increased transcription, post transcriptional processing, translation, post translational processing, altered stability, and altered protein degradation), as well as local overexpression due to altered protein traffic patterns (increased nuclear localization), and augmented functional activity, e.g., as in an increased enzyme hydrolysis of substrate.
  • overexpression refers to either protein or RNA levels. Overexpression can also be by 50%, 60%, 70%, 80%, 90% or more in comparison to a normal cell or comparison cell.
  • the anti-B7-H3 antibodies or ADCs of the invention are used to treat solid tumors likely to overexpress B7-H3
  • the term“gene amplification”, as used herein, refers to a cellular process characterized by the production of multiple copies of any particular piece of DNA.
  • a tumor cell may amplify, or copy, chromosomal segments as a result of cell signals and sometimes environmental events.
  • the process of gene amplification leads to the production of additional copies of the gene.
  • the gene is B7-H3, i.e.,“B7-H3 amplification.”
  • the compositions and methods disclosed herein are used to treat a subject having B7-H3 amplified cancer.
  • administering is meant to refer to the delivery of a substance (e.g., an anti-B7-H3 antibody or ADC) to achieve a therapeutic objective (e.g., the treatment of a B7-H3- associated disorder).
  • Modes of administration may be parenteral, enteral and topical.
  • Parenteral administration is usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.
  • combination therapy or“combination” in the context of a therapeutic method (e.g., a treatment method), refers to the administration of two or more therapeutic substances, e.g., an anti-B7-H3 antibody or ADC and an additional therapeutic agent.
  • the additional therapeutic agent may be administered concomitant with, prior to, or following the administration of the anti-B7-H3 antibody or ADC.
  • the term“effective amount” or“therapeutically effective amount” refers to the amount of a drug, e.g., an antibody or ADC, which is sufficient to reduce or ameliorate the severity and/or duration of a disorder, e.g., cancer, or one or more symptoms thereof, prevent the advancement of a disorder, cause regression of a disorder, prevent the recurrence, development, onset or progression of one or more symptoms associated with a disorder, detect a disorder, or enhance or improve the prophylactic or therapeutic effect(s) of another therapy (e.g., prophylactic or therapeutic agent).
  • a drug e.g., an antibody or ADC
  • the effective amount of an antibody or ADC may, for example, inhibit tumor growth (e.g., inhibit an increase in tumor volume), decrease tumor growth (e.g., decrease tumor volume), reduce the number of cancer cells, and/or relieve to some extent one or more of the symptoms associated with the cancer.
  • the effective amount may, for example, improve disease free survival (DFS), improve overall survival (OS), or decrease likelihood of recurrence.
  • substituents are defined below.
  • the number of carbon atoms in a substituent is indicated by the prefix“C x -C y ” or“C x-y ” wherein x is the minimum and y is the maximum number of carbon atoms.
  • “C 1 -C 6 alkyl” refers to an alkyl containing from 1 to 6 carbon atoms.
  • “C 3 -C 8 cycloalkyl” means a saturated hydrocarbyl ring containing from 3 to 8 carbon ring atoms. If a substituent is described as being“substituted,” a hydrogen atom on a carbon or nitrogen is replaced with a non-hydrogen group.
  • a substituted alkyl substituent is an alkyl substituent in which at least one hydrogen atom on the alkyl is replaced with a non-hydrogen group.
  • monofluoroalkyl is alkyl substituted with a fluoro radical
  • difluoroalkyl is alkyl substituted with two fluoro radicals.
  • substituents include, but are not limited to, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, aryl, cycloalkyl, heterocyclyl, heteroaryl, halogen, C 1 -C 6 haloalkyl, oxo, -CN, NO 2 , -OR xa , -OC(O)R xz , -OC(O)N(R xa ) 2 , -SR xa , -S(O) 2 R xa , -S(O) 2 N(R xa ) 2 , -C(O)R xa , -C(O)OR xa
  • R xa at each occurrence, is independently hydrogen, aryl, cycloalkyl, heterocyclyl, heteroaryl, C 1 -C 6 alkyl, or C 1 -C 6 haloalkyl
  • R xz at each occurrence, is independently aryl, cycloalkyl, heterocyclyl, heteroaryl, C 1 -C 6 alkyl or C 1 -C 6 haloalkyl.
  • alkoxy refers to a group of the formula–OR xa , where R xa ⁇ is an alkyl group.
  • Representative alkoxy groups include methoxy, ethoxy, propoxy, tert-butoxy and the like.
  • alkoxyalkyl refers to an alkyl group substituted with an alkoxy group and may be represented by the general formula–R b OR xa where R b is an alkylene group and R xa is an alkyl group.
  • alkyl by itself or as part of another substituent refers to a saturated or unsaturated branched, straight-chain or cyclic monovalent hydrocarbon radical that is derived by the removal of one hydrogen atom from a single carbon atom of a parent alkane, alkene or alkyne.
  • Typical alkyl groups include, but are not limited to, methyl; ethyls such as ethanyl, ethenyl, ethynyl; propyls such as propan-1-yl, propan-2-yl, cyclopropan-1-yl, prop-1-en-1-yl, prop-1-en-2-yl, prop-2-en-1-yl, cycloprop-1-en-1-yl; cycloprop-2-en-1-yl, prop-1-yn-1-yl , prop-2-yn-1-yl, etc.; butyls such as butan-1-yl butan-2-yl 2-methyl-propan-1-yl 2-methyl-propan-2-yl cyclobutan-1-yl but-1-en-1-yl, but-1-en-2-yl, 2-methyl-prop-1-en-1-yl, but-2-en-1-yl , but-2-en-2-yl, buta-1,3-dien-1
  • alkanyl by itself or as part of another substituent refers to a saturated branched, straight-chain or cyclic alkyl derived by the removal of one hydrogen atom from a single carbon atom of a parent alkane.
  • Typical alkanyl groups include, but are not limited to, methyl; ethanyl; propanyls such as propan-1-yl, propan-2-yl (isopropyl), cyclopropan-1-yl, etc.; butanyls such as butan-1-yl, butan-2-yl (sec-butyl), 2-methyl-propan-1-yl (isobutyl), 2-methyl-propan-2-yl (t-butyl),
  • alkenyl by itself or as part of another substituent refers to an unsaturated branched, straight-chain or cyclic alkyl having at least one carbon-carbon double bond derived by the removal of one hydrogen atom from a single carbon atom of a parent alkene.
  • Typical alkenyl groups include, but are not limited to, ethenyl; propenyls such as prop-1-en-1-yl , prop-1-en-2-yl, prop-2-en-1-yl, prop-2-en-2-yl, cycloprop-1-en-1-yl; cycloprop-2-en-1-yl ; butenyls such as but-1-en-1-yl, but-1-en-2-yl, 2-methyl-prop-1-en-1-yl, but-2-en-1-yl, but-2-en-2-yl,
  • alkynyl by itself or as part of another substituent refers to an unsaturated branched, straight-chain or cyclic alkyl having at least one carbon-carbon triple bond derived by the removal of one hydrogen atom from a single carbon atom of a parent alkyne.
  • Typical alkynyl groups include, but are not limited to, ethynyl; propynyls such as prop-1-yn-1-yl , prop-2-yn-1-yl, etc.;
  • butynyls such as but-1-yn-1-yl, but-1-yn-3-yl, but-3-yn-1-yl , etc.; and the like.
  • alkylamine refers to a group of the formula -NHR xa and“dialkylamine” refers to a group of the formula–NR xa R xa , where each R xa is, independently of the others, an alkyl group.
  • alkylene refers to an alkane, alkene or alkyne group having two terminal monovalent radical centers derived by the removal of one hydrogen atom from each of the two terminal carbon atoms.
  • Typical alkylene groups include, but are not limited to, methylene; and saturated or unsaturated ethylene; propylene; butylene; and the like.
  • lower alkylene refers to alkylene groups with 1 to 6 carbons.
  • heteroalkylene refers to a divalent alkylene having one or more—CH 2 — groups replaced with a thio, oxy, or—NR x3 — where R x3 is selected from hydrogen, lower alkyl and lower heteroalkyl.
  • the heteroalkylene can be linear, branched, cyclic, bicyclic, or a combination thereof and can include up to 10 carbon atoms and up to 4 heteroatoms.
  • heteroalkylene refers to alkylene groups with 1 to 4 carbon atoms and 1 to 3 heteroatoms.
  • aryl means an aromatic carbocyclyl containing from 6 to 14 carbon ring atoms.
  • An aryl may be monocyclic or polycyclic (i.e., may contain more than one ring). In the case of polycyclic aromatic rings, only one ring the polycyclic system is required to be aromatic while the remaining ring(s) may be saturated, partially saturated or unsaturated.
  • aryls include phenyl, naphthalenyl, indenyl, indanyl, and tetrahydronaphthyl.
  • arylene refers to an aryl group having two monovalent radical centers derived by the removal of one hydrogen atom from each of the two ring carbons.
  • An exemplary arylene group is a phenylene.
  • An alkyl group may be substituted by a“carbonyl” which means that two hydrogen atoms from a single alkanylene carbon atom are removed and replaced with a double bond to an oxygen atom.
  • haloalkyl means an alkyl substituent in which at least one hydrogen radical is replaced with a halogen radical.
  • Typical halogen radicals include chloro, fluoro, bromo and iodo.
  • Examples of haloalkyls include chloromethyl, 1- bromoethyl, fluoromethyl, difluoromethyl, trifluoromethyl, and 1,1,1-trifluoroethyl. It should be recognized that if a substituent is substituted by more than one halogen radical, those halogen radicals may be identical or different (unless otherwise stated).
  • haloalkoxy refers to a group of the formula–OR c , where R c is a haloalkyl.
  • R c is a haloalkyl.
  • heteroalkyl refers to alkyl, alkanyl, alkenyl, alkynyl, and alkylene groups, respectively, in which one or more of the carbon atoms, e.g., 1, 2 or 3 carbon atoms, are each independently replaced with the same or different heteroatoms or heteroatomic groups.
  • Typical heteroatoms and/or heteroatomic groups which can replace the carbon atoms include, but are not limited to, -O-, -S-, -S-O-, -NR c -, -PH, -S(O)-, -S(O) 2 -, -S(O)NR c -, -S(O) 2 NR c -, and the like, including combinations thereof, where each R c is independently hydrogen or C 1 -C 6 alkyl.
  • the term“lower heteroalkyl” refers to between 1 and 4 carbon atoms and between 1 and 3 heteroatoms.
  • cycloalkyl and“heterocyclyl” refer to cyclic versions of“alkyl” and “heteroalkyl” groups, respectively.
  • heterocyclyl groups a heteroatom can occupy the position that is attached to the remainder of the molecule.
  • a cycloalkyl or heterocyclyl ring may be a single- ring (monocyclic) or have two or more rings (bicyclic or polycyclic).
  • Monocyclic cycloalkyl and heterocyclyl groups will typically contains from 3 to 7 ring atoms, more typically from 3 to 6 ring atoms, and even more typically 5 to 6 ring atoms.
  • Examples of cycloalkyl groups include, but are not limited to, cyclopropyl; cyclobutyls such as cyclobutanyl and cyclobutenyl; cyclopentyls such as cyclopentanyl and cyclopentenyl; cyclohexyls such as
  • cyclohexanyl and cyclohexenyl examples include, but are not limited to oxetane furanyl dihydrofuranyl tetrahydrofuranyl tetrahydropyranyl thiophenyl (thiofuranyl), dihydrothiophenyl, tetrahydrothiophenyl, pyrrolyl, pyrrolinyl, pyrrolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, triazolyl, tetrazolyl, oxazolyl, oxazolidinyl, isoxazolidinyl, isoxazolidinyl, isoxazolyl, thiazolyl, isothiazolyl, thiazolinyl, isothiazolinyl, thiazolidinyl, isoxazolid
  • Polycyclic cycloalkyl and heterocyclyl groups contain more than one ring, and bicyclic cycloalkyl and heterocyclyl groups contain two rings. The rings may be in a bridged, fused or spiro orientation. Polycyclic cycloalkyl and heterocyclyl groups may include combinations of bridged, fused and/or spiro rings. In a spirocyclic cycloalkyl or heterocyclyl, one atom is common to two different rings.
  • An example of a spirocycloalkyl is spiro[4.5]decane and an example of a
  • spiroheterocyclyls is a spiropyrazoline.
  • bridged cycloalkyl or heterocyclyl the rings share at least two common non-adjacent atoms.
  • bridged cycloalkyls include, but are not limited to, adamantyl and norbornanyl rings.
  • bridged heterocyclyls include, but are not limited to, 2- oxatricyclo[3.3.1.1 3,7 ]decanyl.
  • fused-ring cycloalkyl or heterocyclyl two or more rings are fused together, such that two rings share one common bond.
  • fused-ring cycloalkyls include decalin, naphthylene, tetralin, and anthracene.
  • fused-ring heterocyclyls containing two or three rings include imidazopyrazinyl (including imidazo[1,2-a]pyrazinyl), imidazopyridinyl (including imidazo[1,2- a]pyridinyl), imidazopyridazinyl (including imidazo[1,2-b]pyridazinyl), thiazolopyridinyl (including thiazolo[5,4-c]pyridinyl, thiazolo[5,4-b]pyridinyl, thiazolo[4,5-b]pyridinyl, and thiazolo[4,5- c]pyridinyl), indolizinyl, pyranopyrrolyl, 4H-quinolizinyl, purinyl, naphthyridinyl, pyridopyridinyl (including pyrido[3,4-b]-pyridinyl, pyrido[3,2-b]-pyr
  • fused-ring heterocyclyls include benzo-fused heterocyclyls, such as dihydrochromenyl, tetrahydroisoquinolinyl, indolyl, isoindolyl (isobenzazolyl, pseudoisoindolyl), indoleninyl (pseudoindolyl) isoindazolyl (benzpyrazolyl) benzazinyl (including quinolinyl (1- benzazinyl) or isoquinolinyl (2-benzazinyl)), phthalazinyl, quinoxalinyl, quinazolinyl, benzodiazinyl (including cinnolinyl (1,2-benzodiazinyl) or quinazolinyl (1,3-benzodiazinyl)), benzopyranyl (including chromanyl or isochromanyl), benzoxazinyl (including 1,3,2-benzoxazinyl, 1,
  • heteroaryl refers to an aromatic heterocyclyl containing from 5 to 14 ring atoms.
  • a heteroaryl may be a single ring or 2 or 3 fused rings.
  • heteroaryls include 6- membered rings such as pyridyl, pyrazyl, pyrimidinyl, pyridazinyl, and 1,3,5-, 1,2,4- or 1,2,3- triazinyl; 5-membered ring substituents such as triazolyl, pyrrolyl, imidazyl, furanyl, thiophenyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, 1,2,3-, 1,2,4-, 1,2,5-, or 1,3,4-oxadiazolyl and isothiazolyl; 6/5-membered fused ring substituents such as imidazopyrazinyl (including imidazo[1,2- a]pyrazinyl)imidazo
  • benzothiofuranyl benzisoxazolyl, benzoxazolyl, purinyl, and anthranilyl
  • 6/6-membered fused rings such as benzopyranyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, and benzoxazinyl.
  • Heteroaryls may also be heterocycles having aromatic (4N+2 pi electron) resonance contributors such as pyridonyl (including pyrid-2(1H)-onyl and pyrid-4(1H)-onyl), pyrimidonyl (including pyramid- 2(1H)-onyl and pyramid-4(3H)-onyl), pyridazin-3(2H)-onyl and pyrazin-2(1H)-onyl.
  • aromatic (4N+2 pi electron) resonance contributors such as pyridonyl (including pyrid-2(1H)-onyl and pyrid-4(1H)-onyl), pyrimidonyl (including pyramid- 2(1H)-onyl and pyramid-4(3H)-onyl), pyridazin-3(2H)-onyl and pyrazin-2(1H)-onyl.
  • sulfonate as used herein means a salt or ester of a sulfonic acid.
  • methyl sulfonate as used herein means a methyl ester of a sulfonic acid group.
  • carboxylate as used herein means a salt or ester of a carboxylic acid.
  • polyol as used herein, means a group containing more than two hydroxyl groups independently or as a portion of a monomer unit. Polyols include, but are not limited to, reduced C 2 -C 6 carbohydrates, ethylene glycol, and glycerin.
  • “sugar” when used in context of“G 1 ” includes O-glycoside, N-glycoside, S- glycoside and C-glycoside (C-glycoslyl) carbohydrate derivatives of the monosaccharide and disaccharide classes and may originate from naturally-occurring sources or may be synthetic in origin.
  • “sugar” when used in context of“G 1 ” includes derivatives such as but not limited to those derived from glucuronic acid, galacturonic acid, galactose, and glucose among others. Suitable sugar substitutions include but are not limited to hydroxyl, amine, carboxylic acid, sulfonic acid, phosphonic acid, esters, and ethers.
  • N-hydroxysuccinimide ester derivative of a carboxylic acid means the N-hydroxysuccinimide ester derivative of a carboxylic acid.
  • amine includes primary, secondary and tertiary aliphatic amines, including cyclic versions.
  • salt when used in context of“or salt thereof” include salts commonly used to form alkali metal salts and to form addition salts of free acids or free bases. In general, these salts typically may be prepared by conventional means by reacting, for example, the appropriate acid or base with a compound of the invention
  • the salt preferably is pharmaceutically acceptable and/or physiologically compatible.
  • pharmaceutically acceptable is used adjectivally in this patent application to mean that the modified noun is appropriate for use as a pharmaceutical product or as a part of a pharmaceutical product.
  • pharmaceutically acceptable salt includes salts commonly used to form alkali metal salts and to form addition salts of free acids or free bases. In general, these salts typically may be prepared by conventional means by reacting, for example, the appropriate acid or base with a compound of the invention.
  • ADCs antibody drug conjugates comprising an anti-B7-H3 antibody described herein and at least one drug(s), such as, but not limited to, a Bcl-xL inhibitor or an auristatin.
  • the antibodies or ADCs of the invention have characteristics including, but not limited to, binding to wild- type human B7-H3 in vitro, binding to wild-type human B7-H3 on tumor cells expressing B7-H3, and decreasing or inhibiting xenograft tumor growth in a mouse model.
  • One aspect of the invention features an anti-human B7-H3 (anti-hB7-H3) Antibody Drug Conjugate (ADC) comprising an anti-hB7-H3 antibody conjugated to a drug via a linker, wherein the drug is a Bcl-xL inhibitor.
  • ADC Antibody Drug Conjugate
  • the anti-B7-H3 antibodies described herein provide the ADCs of the invention with the ability to bind to B7-H3 such that the cytotoxic Bcl-xL drug attached to the antibody may be delivered to the B7-H3-expressing cell, particularly a B7-H3 expressing cancer cell.
  • antibody fragments i.e., antigen-binding portions of an anti-B7-H3antibody
  • an anti-B7-H3antibody fragment may be conjugated to the Bcl-xL inhibitors described herein.
  • antibody fragments of the anti-B7- H3antibodies described herein are conjugated to Bcl-xL inhibitors (including those described below in Section III.A) via linkers (including those described below in Section III.A).
  • the anti-B7-H3 antibody binding portion is a Fab, a Fab’, a F(ab’)2, a Fv, a disulfide linked Fv, an scFv, a single domain antibody, or a diabody.
  • a chimeric antibody is a molecule in which different portions of the antibody are derived from different animal species, such as antibodies having a variable region derived from a murine monoclonal antibody and a human immunoglobulin constant region.
  • Methods for producing chimeric antibodies are known in the art. See e.g., Morrison, Science 229:1202 (1985); Oi et al., BioTechniques 4:214 (1986); Gillies et al., (1989) J. Immunol. Methods 125:191-202; U.S. Pat. Nos.5,807,715; 4,816,567; and 4,816,397, which are incorporated herein by reference in their entireties.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence set forth in SEQ ID NOs: 1, 9, 16, 24, 32, 40, 48, 56, 64, 72, 80, 87, 95, 101, or 108; and/or a light chain variable region including an amino acid sequence set forth in SEQ ID NOs: 5, 13, 20, 28, 36, 44, 52, 60, 68, 76, 84, 91, 98, 105, or 112.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 1, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 5.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 2; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 3; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 4; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 6; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 8.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 9, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 13.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 11; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 14 (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 16, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 20.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 17; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 18; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 19; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 21; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 22; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 23.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 24, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 28.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 25; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 26; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 27; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 29; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 30; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 31.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 32, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 36.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 33; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 34; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 35; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 37; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 38; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 182.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 40, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 44.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 41; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 42; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 43; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 45; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 46; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 47.
  • a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 41; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 42; and (c) a CDR3
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 48, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 52.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 49; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 50; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 51; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 53; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 54; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 55.
  • a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 49; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 50; and (c) a CDR3
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 56, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 60.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 57; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 58; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 59; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 61; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 62; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 63.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 64, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 68.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 65; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 66; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 67; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 69; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 70; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 71.
  • a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 65; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 66; and (c)
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 72, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 76.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 73; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 74; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 75; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 77; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 78; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 79.
  • a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 73; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 74; and (
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 80, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 84.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 81; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 82; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 83; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 85; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 86.
  • a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 81; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 82; and (c
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 87 and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 91.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 88; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 89; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 90; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 92; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 93; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 94.
  • a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 88; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 89; and (
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 95, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 98.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 49; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 96; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 97; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 99; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 93; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 100.
  • a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 49; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 96; and (c)
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 101, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 105.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 102; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 103; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 104; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 106; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 46; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 107.
  • a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 102; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 103; and (
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 108, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 112.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 109; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 110; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 111; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 113; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 114; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 115.
  • a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 109; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 110; and (c
  • the chimeric antibodies disclosed herein may be used in the production of humanized anti- B7-H3 antibodies. For example, following the generation and characterization of chimeric anti-B7- H3 antibodies chAb1-chAb18, antibodies chAb3, chAb13, and chAb18 were selected for
  • huAb3v1 six different humanized antibodies were created based on chAb3 (referred to herein as huAb3v1, huAb3v2, huAb3v3, huAb3v4, huAb3v5, and huAb3v6 (see
  • chAb13 nine different humanized antibodies were created based on chAb13 (referred to herein as huAb13v1, huAb13v2, huAb13v3, huAb13v4, huAb13v5, huAb13v6, huAb13v7, huAb13v8, huAb13v9), and ten different humanized antibodies were created based on chAb18 (referred to herein as huAb18v1, huAb18v2, huAb18v3, huAb18v4, huAb18v5, huAb18v6, huAb18v7, huAb18v8, huAb18v9, and huAb18v10 (see Examples 9 and 10)).
  • Tables 8, 12, 16, 18, and 19 provide the amino acid sequences of CDR, VH and VL regions of humanized chAb3, chAb13, and chAb18, respectively.
  • humanized antibodies are antibody molecules from non-human species antibody that binds the desired antigen having one or more complementarity determining regions (CDRs) from the non-human species and framework regions from a human immunoglobulin molecule.
  • CDRs complementarity determining regions
  • Known human Ig sequences are disclosed, e.g., www.ncbi.nlm.nih.gov/entrez- /query.fcgi;
  • Framework residues in the human framework regions may be substituted with the corresponding residue from the CDR donor antibody to alter, preferably improve, antigen binding.
  • These framework substitutions are identified by methods well known in the art, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions. (See, e.g., Queen et al., U.S. Pat. No.5,585,089; Riechmann et al., Nature 332:323 (1988), which are incorporated herein by reference in their entireties.) Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art.
  • Antibodies can be humanized using a variety of techniques known in the art, such as but not limited to those described in Jones et al., Nature 321:522 (1986); Verhoeyen et al., Science 239:1534 (1988)), Sims et al., J. Immunol.151: 2296 (1993); Chothia and Lesk, J. Mol. Biol.196:901 (1987), Carter et al., Proc. Natl. Acad. Sci. U.S.A.89:4285 (1992); Presta et al., J.
  • huAb3v1 VH amino acid sequence set forth in SEQ ID NO: 125 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 11, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 128 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 14, 7, and 15, respectively;
  • huAb3v2 VH amino acid sequence set forth in SEQ ID NO: 127 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 11, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 128 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 14, 7, and 15, respectively;
  • huAb3v3 VH amino acid sequence set forth in SEQ ID NO: 126 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 11, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 129 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 14, 7, and 15, respectively;
  • huAb3v4 VH amino acid sequence set forth in SEQ ID NO: 125 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 11, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 130 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 14, 7, and 15, respectively;
  • huAb3v5 VH amino acid sequence set forth in SEQ ID NO: 127 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 11, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 130 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 14, 7, and 15, respectively; and
  • huAb3v6 VH amino acid sequence set forth in SEQ ID NO: 126 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 11, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 130 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 14, 7, and 15, respectively.
  • huAb3v2 was selected for further modified in order to remove potential deamidation or isomerization sites in the light chain CDR1 or in the heavy chain CDR2.
  • huAb3v2 Nine variants of the humanized antibody huAb3v2 were generated, and are referred to herein as huAb3v2.1, huAb3v2.2, huAb3v2.3, huAb3v2.4, huAb3v2.5, huAb3v2.6, huAb3v2.7, huAb3v2.8, and huAb3v2.9 (CDR and variable domain sequences are provided in Table 13).
  • the nine variants of the huAb3v2 antibody include the following:
  • huAb3v2.1 VH amino acid sequence set forth in SEQ ID NO: 131 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 132, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 133 and VL CDR1 CDR2 and CDR3 amino acid sequences set forth in SEQ ID NOs: 134, 7, and 15, respectively;
  • huAb3v2.2 VH amino acid sequence set forth in SEQ ID NO: 131 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 132, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 135 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 136, 7, and 15, respectively;
  • huAb3v2.4 VH amino acid sequence set forth in SEQ ID NO: 139 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 140, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 133 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 134, 7, and 15, respectively;
  • huAb3v2.5 VH amino acid sequence set forth in SEQ ID NO: 139 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 140, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 135 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 136, 7, and 15, respectively;
  • huAb3v2.6 VH amino acid sequence set forth in SEQ ID NO: 139 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 140, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 137 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 138, 7, and 15, respectively;
  • H huAb3v2.8 (VH amino acid sequence set forth in SEQ ID NO: 141 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 142, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 135 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 136, 7, and 15, respectively); and
  • huAb3v2.9 VH amino acid sequence set forth in SEQ ID NO: 141 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 142, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 137 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 138, 7, and 15, respectively.
  • the present invention provides antibodies comprising variable and/or CDR sequences from a humanized antibody derived from chAb3.
  • the invention features anti-B7-H3 antibodies which are derived from Ab3 have improved characteristics, e.g., improved binding affinity to isolated B7-H3 protein and improved binding to B7-H3 expressing cells as described in the Examples below. Collectively these novel antibodies are referred to herein as “Ab3 variant antibodies.” Generally, the Ab3 variant antibodies retain the same epitope specificity as Ab3.
  • anti-B7-H3 antibodies, or antigen binding fragments thereof, of the invention are capable of modulating a biological function of B7-H3.
  • the present invention provides a humanized antibody, or antigen binding portion thereof, having a heavy chain variable region including an amino acid sequence set forth in SEQ ID NOs: 125, 126, 127, 131, 139, or 141; and/or a light chain variable region including an amino acid sequence set forth in SEQ ID NOs: 128, 129, 130, 133, 135, or 137.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen binding portion thereof, of the invention comprises a heavy chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 10; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 11, 132, 140, or 142; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 14, 134, 136, or 138; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 7; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 15.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 125, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 128.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 127, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 128.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 126, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 129.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 125, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 130.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 127, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 130.
  • the present invention is directed to an anti-B7-H3 antibody or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 126, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 130.
  • the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 11; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 14; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 131, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 133.
  • the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 132; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 134; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 131, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 135.
  • the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 132; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 136; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 131, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 137.
  • the present invention is directed to a humanized anti-B7-H3 antibody or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 132; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 138; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 139, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 133.
  • the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 140; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 134; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 139, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 135.
  • the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 140; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 136; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain comprising the amino acid sequence of SEQ ID NO: 170 and a light chain comprising the amino acid sequence of SEQ ID NO: 171.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 139, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 137.
  • the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 140; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 138; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain comprising the amino acid sequence of SEQ ID NO: 172 and a light chain comprising the amino acid sequence of SEQ ID NO: 173.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 141, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 133
  • the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 142; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 134; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 141, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 135.
  • the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 142; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 136; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 141, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 137.
  • the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 142; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 138; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
  • the nine different humanized antibodies created based on chAb13 include the following: A) huAb13v1 (VH amino acid sequence set forth in SEQ ID NO: 147 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 33, 34, and 35, respectively; and VL amino acid sequence set forth in SEQ ID NO: 144 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 37, 38, and 39, respectively);
  • huAb13v2 VH amino acid sequence set forth in SEQ ID NO: 146 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 33, 34, and 35, respectively; and VL amino acid sequence set forth in SEQ ID NO: 143 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 37, 38, and 39, respectively;
  • huAb13v3 VH amino acid sequence set forth in SEQ ID NO: 146 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 33, 34, and 35, respectively; and VL amino acid sequence set forth in SEQ ID NO: 144 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 37, 38, and 39, respectively;
  • huAb13v4 VH amino acid sequence set forth in SEQ ID NO: 146 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 33, 34, and 35, respectively; and VL amino acid sequence set forth in SEQ ID NO: 145 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 37, 38, and 39, respectively;
  • huAb13v5 VH amino acid sequence set forth in SEQ ID NO: 147 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 33, 34, and 35, respectively; and VL amino acid sequence set forth in SEQ ID NO: 143 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 37, 38, and 39, respectively;
  • huAb13v6 VH amino acid sequence set forth in SEQ ID NO: 147 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 33, 34, and 35, respectively; and VL amino acid sequence set forth in SEQ ID NO: 145 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 37, 38, and 39, respectively;
  • huAb13v7 VH amino acid sequence set forth in SEQ ID NO: 148 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 33, 34, and 35, respectively; and VL amino acid sequence set forth in SEQ ID NO: 143 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 37, 38, and 39, respectively;
  • H huAb13v8 (VH amino acid sequence set forth in SEQ ID NO: 148 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 33, 34, and 35, respectively; and VL amino acid sequence set forth in SEQ ID NO: 144 and VL CDR1 CDR2 and CDR3 amino acid sequences set forth in SEQ ID NOs: 37, 38, and 39, respectively);
  • huAb13v9 VH amino acid sequence set forth in SEQ ID NO: 148 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 33, 34, and 35, respectively; and VL amino acid sequence set forth in SEQ ID NO: 145 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 37, 38, and 39, respectively.
  • the present invention provides antibodies comprising variable and/or CDR sequences from a humanized antibody derived from chAb13.
  • the invention features anti-B7-H3 antibodies which are derived from chAb13 have improved characteristics, e.g., improved binding affinity to isolated B7-H3 protein and improved binding to B7-H3 expressing cells, as described in the Examples below. Collectively these novel antibodies are referred to herein as “Ab13 variant antibodies.” Generally, the Ab13 variant antibodies retain the same epitope specificity as Ab13.
  • anti-B7-H3 antibodies, or antigen binding fragments thereof, of the invention are capable of modulating a biological function of B7-H3.
  • the present invention provides a humanized antibody, or antigen binding portion thereof, having a heavy chain variable region including an amino acid sequence set forth in SEQ ID NOs: 146, 147, or 148; and/or a light chain variable region including an amino acid sequence set forth in SEQ ID NOs: 143, 144, or 145.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen binding portion thereof, of the invention comprises a heavy chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 33; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 34; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 35; and a light chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 37; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 38; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 39.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 147, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 144.
  • the invention provides an anti-B7H3 antibody comprising the CDR sequences set forth in the variable regions of huAb13v1 (SEQ ID NOs.144 and 147).
  • the present invention is directed to an anti-B7-H3 antibody, or antigen binding portion thereof, having a heavy chain comprising the amino acid sequence of SEQ ID NO: 168 and a light chain comprising the amino acid sequence of SEQ ID NO: 169.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 146, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 143
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 146, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 144.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 146, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 145.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 147, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 143.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 147, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 145.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 148, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 143.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 148, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 144.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 148, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 145.
  • Humanized anti-B7-H3 antibodies derived from chAb18 having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 148, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 145.
  • the ten different humanized antibodies created based on chAb18 include the following: A) huAb18v1 (VH amino acid sequence set forth in SEQ ID NO: 116 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25, 26, and 27, respectively; and VL amino acid sequence set forth in SEQ ID NO: 120 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively);
  • huAb18v2 VH amino acid sequence set forth in SEQ ID NO: 118 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25 119 and 27 respectively; and VL amino acid sequence set forth in SEQ ID NO: 120 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively;
  • huAb18v3 VH amino acid sequence set forth in SEQ ID NO: 117 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25, 26, and 27, respectively; and VL amino acid sequence set forth in SEQ ID NO: 121 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively;
  • huAb18v4 VH amino acid sequence set forth in SEQ ID NO: 118 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25, 119, and 27, respectively; and VL amino acid sequence set forth in SEQ ID NO: 121 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively;
  • huAb18v5 VH amino acid sequence set forth in SEQ ID NO: 116 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25, 26, and 27, respectively; and VL amino acid sequence set forth in SEQ ID NO: 123 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively;
  • huAb18v6 VH amino acid sequence set forth in SEQ ID NO: 118 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25, 119, and 27, respectively; and VL amino acid sequence set forth in SEQ ID NO: 123 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively;
  • huAb18v7 VH amino acid sequence set forth in SEQ ID NO: 118 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25, 119, and 27, respectively; and VL amino acid sequence set forth in SEQ ID NO: 124 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively;
  • H huAb18v8 (VH amino acid sequence set forth in SEQ ID NO: 117 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25, 26, and 27, respectively; and VL amino acid sequence set forth in SEQ ID NO: 122 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively);
  • huAb18v9 VH amino acid sequence set forth in SEQ ID NO: 117 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25, 26, and 27, respectively; and VL amino acid sequence set forth in SEQ ID NO: 124 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively; and
  • huAb18v10 VH amino acid sequence set forth in SEQ ID NO: 118 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25, 119, and 27, respectively; and VL amino acid sequence set forth in SEQ ID NO: 122 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively.
  • the present invention provides antibodies comprising variable and/or CDR sequences from a humanized antibody derived from chAb18.
  • the invention features anti-B7-H3 antibodies which are derived from Ab18 have improved characteristics e g improved binding affinity to isolated B7-H3 protein and improved binding to B7-H3 expressing cells, as described in the Examples below. Collectively these novel antibodies are referred to herein as “Ab18 variant antibodies.” Generally, the Ab18 variant antibodies retain the same epitope specificity as Ab18.
  • anti-B7-H3 antibodies, or antigen binding fragments thereof, of the invention are capable of modulating a biological function of B7-H3.
  • the present invention provides a humanized antibody, or antigen binding portion thereof, having a heavy chain variable region including an amino acid sequence set forth in SEQ ID NOs: 116, 117, or 118; and/or a light chain variable region including an amino acid sequence set forth in SEQ ID NOs: 120, 121, 122, 123 or 124.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen binding portion thereof, of the invention comprises a heavy chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 25; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 26 or 119; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 27; and a light chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 29; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 30; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 31.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 116, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 120.
  • the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 25; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 26; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 27; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 29; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 30; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 31.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 118, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 120.
  • the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 25; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 119; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 27; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 29; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 30; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 31.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 117, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 121.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 118, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 121.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 116, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 123.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 118, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 123.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 118, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 124.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 117, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 122.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 117, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 124.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 118, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 122.
  • the present invention provides a humanized antibody, or antigen binding portion thereof, having a heavy chain variable region including an amino acid sequence set forth in SEQ ID NOs: 116 117 118 146 147 148 125 126 127 131 139 or 141; and/or a light chain variable region including an amino acid sequence set forth in SEQ ID NOs: 120, 121, 122, 123, 124, 143, 144, 145, 128, 129, 130, 133, 135, or 137.
  • the present invention is directed to an anti-B7-H3 antibody, or antigen binding portion thereof, of the invention comprises a heavy chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 10, 25, or 33; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 11, 26, 34, 119, 132, 140, or 142; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 12, 27, or 35; and a light chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 14, 29, 37, 134, 136, or 138; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 7, 30, or 38; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 15, 31 or 39.
  • the invention provides an anti-B7-H3 antibody, or antigen binding fragment thereof, that specifically competes with an anti-B7-H3 antibody, or fragment thereof, described herein, wherein said competition can be detected in a competitive binding assay using said antibody, the human B7-H3 polypeptide, and the anti-B7-H3 antibody or fragment thereof.
  • the competing antibody, or antigen binding portion thereof is an antibody, or antigen binding portion thereof, that competes with huAb3v2.5, huAb3v2.6, or huAb13v1.
  • the anti-B7-H3 antibodies, or antigen binding portions thereof, of the invention bind to the extracellular domain of human B7-H3 (SEQ ID NO: 152) with a dissociation constant (K D ) of about 1 x 10 -6 M or less, as determined by surface plasmon resonance.
  • the antibodies, or antigen binding portions thereof bind to human B7-H3 with a K D of between about 1 x 10 -6 M and about 1 x 10 -11 M, as determined by surface plasmon resonance.
  • antibodies, or antigen binding portions thereof bind to human B7-H3 with a K D of between about 1 x 10 -6 M and about 1 x 10 -7 M, as determined by surface plasmon resonance.
  • antibodies, or antigen binding portions thereof, of the invention binds to human B7-H3 with a K D of between about 1 x 10 -6 M and about 5 x 10 -11 M, about 1 x 10 -6 M and about 5 x 10 -10 M; a K D of between about 1 x 10 -6 M and about 1 x 10 -9 M; a K D of between about 1 x 10 -6 M and about 5 x 10 -9 M; a K D of between about 1 x 10 -6 M and about 1 x 10 -8 M; a K D of between about 1 x 10 -6 M and about 5 x 10 -8 M; a K D of between about 8.4 x 10 -7 M and about 3.4 x 10 -11 M; a K D of between about 5.9 x 10 -7 M; and about 2.2 x 10 -7 M, as determined by surface plasmon resonance.
  • the antibodies, or antigen binding portions thereof, of the invention bind to human B7-H3 (SEQ ID NO: 149) with a K D of about 1 x 10 -6 M or less, as determined by surface plasmon resonance.
  • the antibodies, or antigen binding portions thereof, of the invention bind to human B7-H3 (SEQ ID NO: 149) with a K D of between about 8.2 x 10 -9 M and about 6.3 x 10- 10 M; a K D of between about 8.2 x 10 -9 M and about 2.0 x 10 -9 M; a K D of between about 2.3 x 10 -9 M and about 1.5 x 10 -10 M, as determined by surface plasmon resonance.
  • Anti-B7-H3 antibodies provided herein may comprise a heavy chain variable region comprising CDR1, CDR2 and CDR3 sequences and a light chain variable region comprising CDR1, CDR2 and CDR3 sequences, wherein one or more of these CDR sequences comprise specified amino acid sequences based on the antibodies described herein (e.g., huAb13v1 or huAb3v2.5), or conservative modifications thereof, and wherein the antibodies retain the desired functional properties of the anti-B7-H3antibodies described herein.
  • the anti-B7-H3 antibody, or antigen binding portion thereof may comprise a heavy chain variable region comprising CDR1, CDR2, and CDR3 sequences and a light chain variable region comprising CDR1, CDR2, and CDR3 sequences, wherein: (a) the heavy chain variable region CDR3 sequence comprises SEQ ID NO: 12 or 35, and conservative modifications thereof, e.g., 1, 2, 3, 4, 5, 1-2, 1-3, 1-4 or 1-5 conservative amino acid substitutions; (b) the light chain variable region CDR3 sequence comprises SEQ ID NO: 15 or 39, and conservative modifications thereof, e.g., 1, 2, 3, 4, 5, 1-2, 1-3, 1-4 or 1-5 conservative amino acid substitutions; (c) the antibody specifically binds to B7-H3, and (d) the antibody exhibits 1, 2, 3, 4, 5, 6, or all of the following functional properties described herein, e.g., binding to soluble human B7-H3.
  • the heavy chain variable region CDR3 sequence comprises SEQ ID NO: 12 or 35, and conservative modifications thereof
  • the heavy chain variable region CDR2 sequence comprises SEQ ID NO: 140 or 34, and conservative modifications thereof, e.g., 1, 2, 3, 4, 5, 1-2, 1-3, 1-4 or 1-5 conservative amino acid substitutions; and the light chain variable region CDR2 sequence comprises SEQ ID NO: 7 or 38, and conservative modifications thereof, e.g., 1, 2, 3, 4, 5, 1-2, 1-3, 1-4 or 1-5 conservative amino acid substitutions.
  • the heavy chain variable region CDR1 sequence comprises SEQ ID NO: 10 or 33, and conservative modifications thereof, e.g., 1, 2, 3, 4, 5, 1-2, 1-3, 1-4 or 1-5 conservative amino acid substitutions; and the light chain variable region CDR1 sequence comprises SEQ ID NO: 136, 138, or 37, and conservative modifications thereof, e.g., 1, 2, 3, 4, 5, 1-2, 1-3, 1-4 or 1-5 conservative amino acid substitutions.
  • Conservative amino acid substitutions may also be made in portions of the antibodies other than, or in addition to, the CDRs.
  • conservative amino acid modifications may be made in a framework region or in the Fc region.
  • a variable region or a heavy or light chain may comprise 1, 2, 3, 4, 5, 1-2, 1-3, 1-4, 1-5, 1-10, 1-15, 1-20, 1-25, or 1-50 conservative amino acid substitutions relative to the anti-B7-H3 antibody sequences provided herein.
  • the anti-B7- H3 antibody comprises a combination of conservative and non-conservative amino acid modification.
  • the antibody comprises a heavy chain constant region, such as an IgG1, IgG2, IgG3, IgG4, IgA, IgE, IgM, or IgD constant region.
  • the anti-B7- H3 antibody, or antigen binding portion thereof comprises a heavy chain immunoglobulin constant domain selected from the group consisting of a human IgG constant domain, a human IgM constant domain, a human IgE constant domain, and a human IgA constant domain.
  • the antibody, or antigen binding portion thereof has an IgG1 heavy chain constant region, an IgG2 heavy chain constant region, an IgG3 constant region, or an IgG4 heavy chain constant region.
  • the heavy chain constant region is an IgG1 heavy chain constant region or an IgG4 heavy chain constant region.
  • the antibody can comprise a light chain constant region, either a kappa light chain constant region or a lambda light chain constant region.
  • the antibody comprises a kappa light chain constant region.
  • the antibody portion can be, for example, a Fab fragment or a single chain Fv fragment.
  • the anti-B7-H3 antibody binding portion is a Fab, a Fab’, a F(ab’)2, a Fv, a disulfide linked Fv, an scFv, a single domain antibody, or a diabody.
  • the anti-B7-H3 antibody, or antigen binding portion thereof is a multispecific antibody, e.g. a bispecific antibody.
  • the Fc portion of an antibody mediates several important effector functions e.g. cytokine induction, ADCC, phagocytosis, complement dependent cytotoxicity (CDC) and half-life/ clearance rate of antibody and antigen-antibody complexes. In some cases these effector functions are desirable for therapeutic antibody but in other cases might be unnecessary or even deleterious, depending on the therapeutic objectives.
  • Neonatal Fc receptors are the critical components determining the circulating half-life of antibodies.
  • at least one amino acid residue is replaced in the constant region of the antibody, for example the Fc region of the antibody, such that effector functions of the antibody are altered.
  • One embodiment of the invention includes a recombinant chimeric antigen receptor (CAR) comprising the binding regions of the antibodies described herein, e.g., the heavy and/or light chain CDRs of huAb13v1.
  • a recombinant CAR, as described herein, may be used to redirect T cell specificity to an antigen in a human leukocyte antigen (HLA)-independent fashion.
  • CARs of the invention may be used in immunotherapy to help engineer a human subject’s own immune cells to recognize and attack the subject’s tumor (see, e.g., U.S. Pat.
  • An anti-B7-H3 CAR of the invention preferably contains a extracellular antigen-binding domain specific for B7-H3, a transmembrane domain which is used to anchor the CAR into a T cell, and one or more intracellular signaling domains.
  • the CAR includes a transmembrane domain that comprises a transmembrane domain of a protein selected from the group consisting of the alpha, beta or zeta chain of the T-cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137 and CD154.
  • the CAR comprises a costimulatory domain, e.g., a costimulatory domain comprising a functional signaling domain of a protein selected from the group consisting of OX40, CD2, CD27, CD28, CD5, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), and 4-1BB (CD137).
  • a costimulatory domain comprising a functional signaling domain of a protein selected from the group consisting of OX40, CD2, CD27, CD28, CD5, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), and 4-1BB (CD137).
  • the CAR comprises an scFv comprising the CDR or variable regions described herein e.g., CDRs or variable regions from the huAb13v1 antibody, a transmembrane domain, a co-stimulatory domain (e.g., a functional signaling domain from CD28 or 4-1BB), and a signaling domain comprising a functional signaling domain from CD3 (e.g., CD3-zeta).
  • scFv comprising the CDR or variable regions described herein e.g., CDRs or variable regions from the huAb13v1 antibody, a transmembrane domain, a co-stimulatory domain (e.g., a functional signaling domain from CD28 or 4-1BB), and a signaling domain comprising a functional signaling domain from CD3 (e.g., CD3-zeta).
  • the invention incudes a T cell comprising a CAR (also referred to as a CAR T cell) comprising antigen binding regions, e.g. CDRs, of the antibodies described herein or an scFv described herein.
  • a CAR also referred to as a CAR T cell
  • antigen binding regions e.g. CDRs, of the antibodies described herein or an scFv described herein.
  • the CAR comprises a heavy chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 10, 25, or 33; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 11, 26, 34, 119, 132, 140, or 142; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 12, 27, or 35; and a light chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 14, 29, 37, 134, 136, or 138; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 7, 30, or 38; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 15, 31 or 39.
  • the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 11; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 14; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
  • the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 132; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 134; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
  • the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 132; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 136; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
  • the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 132; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 138; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
  • the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 140; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 134; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
  • the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 140; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 136; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15
  • the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 140; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15
  • the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 142; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 134; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
  • the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 142; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 136; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
  • the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 142; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 138; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
  • the CAR comprises a heavy chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 33; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 34; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 35; and a light chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 37; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 38; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 39.
  • the CAR comprises a heavy chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 25; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 26 or 119; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 27; and a light chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 29; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 30; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 31.
  • the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 25; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 26; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 27; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 29; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 30; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 31.
  • the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 25; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 119; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 27; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 29; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 30; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 31.
  • One embodiment of the invention includes a labeled anti-B7-H3 antibody, or antibody portion thereof, where the antibody is derivatized or linked to one or more functional molecule(s) (e.g., another peptide or protein).
  • a labeled antibody can be derived by functionally linking an antibody or antibody portion of the invention (by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as another antibody (e.g., a bispecific antibody or a diabody), a detectable agent, a pharmaceutical agent, a protein or peptide that can mediate the association of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag), and/or a cytotoxic or therapeutic agent selected from the group consisting of a mitotic inhibitor, an antitumor antibiotic, an immunomodulating agent, a vector for gene therapy, an alkylating agent, an antiangiogenic agent, an antimetabolite, a boro
  • Useful detectable agents with which an antibody or antibody portion thereof, may be derivatized include fluorescent compounds.
  • Exemplary fluorescent detectable agents include fluorescein, fluorescein isothiocyanate, rhodamine, 5-dimethylamine-1-napthalenesulfonyl chloride, phycoerythrin and the like.
  • An antibody may also be derivatized with detectable enzymes, such as alkaline phosphatase, horseradish peroxidase, glucose oxidase and the like. When an antibody is derivatized with a detectable enzyme it is detected by adding additional reagents that the enzyme uses to produce a detectable reaction product.
  • the detectable agent horseradish peroxidase when the detectable agent horseradish peroxidase is present the addition of hydrogen peroxide and diaminobenzidine leads to a colored reaction product, which is detectable.
  • An antibody may also be derivatized with biotin, and detected through indirect measurement of avidin or streptavidin binding.
  • the antibody of the invention is conjugated to an imaging agent.
  • imaging agents examples include, but are not limited to, a radiolabel (e.g., indium), an enzyme, a fluorescent label, a luminescent label, a bioluminescent label, a magnetic label, and biotin.
  • the antibodies or ADCs are linked to a radiolabel, such as, but not limited to, indium ( 111 In).
  • 111 Indium may be used to label the antibodies and ADCs described herein for use in identifying B7-H3 positive tumors.
  • anti-B7-H3 antibodies (or ADCs) described herein are labeled with 111 I via a bifunctional chelator which is a bifunctional cyclohexyl diethylenetriaminepentaacetic acid (DTPA) chelate (see US Patent Nos.5,124,471; 5,434,287; and 5,286,850, each of which is incorporated herein by reference).
  • DTPA bifunctional cyclohexyl diethylenetriaminepentaacetic acid
  • Another embodiment of the invention provides a glycosylated binding protein wherein the anti-B7-H3 antibody or antigen binding portion thereof comprises one or more carbohydrate residues.
  • Nascent in vivo protein production may undergo further processing, known as post-translational modification.
  • sugar (glycosyl) residues may be added enzymatically, a process known as glycosylation.
  • glycosylation The resulting proteins bearing covalently linked oligosaccharide side chains are known as glycosylated proteins or glycoproteins.
  • Antibodies are glycoproteins with one or more carbohydrate residues in the Fc domain, as well as the variable domain.
  • Carbohydrate residues in the Fc domain have important effect on the effector function of the Fc domain, with minimal effect on antigen binding or half-life of the antibody (R. Jefferis, Biotechnol. Prog.21 (2005), pp.11–16).
  • glycosylation of the variable domain may have an effect on the antigen binding activity of the antibody.
  • Glycosylation in the variable domain may have a negative effect on antibody binding affinity, likely due to steric hindrance (Co, M.S., et al., Mol. Immunol. (1993) 30:1361- 1367), or result in increased affinity for the antigen (Wallick, S.C., et al., Exp. Med. (1988) 168:1099-1109; Wright, A., et al., EMBO J. (1991) 10:2717-2723).
  • One aspect of the invention is directed to generating glycosylation site mutants in which the O- or N-linked glycosylation site of the binding protein has been mutated.
  • One skilled in the art can generate such mutants using standard well-known technologies.
  • Glycosylation site mutants that retain the biological activity, but have increased or decreased binding activity, are another object of the invention.
  • the glycosylation of the anti-B7-H3 antibody or antigen binding portion of the invention is modified.
  • an aglycoslated antibody can be made (i.e., the antibody lacks glycosylation).
  • Glycosylation can be altered to, for example, increase the affinity of the antibody for antigen
  • Such carbohydrate modifications can be accomplished by for example altering one or more sites of glycosylation within the antibody sequence.
  • one or more amino acid substitutions can be made that result in elimination of one or more variable region glycosylation sites to thereby eliminate glycosylation at that site.
  • Such aglycosylation may increase the affinity of the antibody for antigen.
  • Such an approach is described in further detail in PCT Publication WO2003016466A2, and U.S. Pat. Nos.5,714,350 and 6,350,861, each of which is incorporated herein by reference in its entirety.
  • a modified anti-B7-H3 antibody of the invention can be made that has an altered type of glycosylation, such as a hypofucosylated antibody having reduced amounts of fucosyl residues or an antibody having increased bisecting GlcNAc structures.
  • altered glycosylation patterns have been demonstrated to increase the ADCC ability of antibodies.
  • carbohydrate modifications can be accomplished by, for example, expressing the antibody in a host cell with altered glycosylation machinery. Cells with altered glycosylation machinery have been described in the art and can be used as host cells in which to express recombinant antibodies of the invention to thereby produce an antibody with altered glycosylation. See, for example, Shields, R. L. et al. (2002) J. Biol.
  • Protein glycosylation depends on the amino acid sequence of the protein of interest, as well as the host cell in which the protein is expressed. Different organisms may produce different glycosylation enzymes (e.g., glycosyltransferases and glycosidases), and have different substrates (nucleotide sugars) available. Due to such factors, protein glycosylation pattern, and composition of glycosyl residues, may differ depending on the host system in which the particular protein is expressed. Glycosyl residues useful in the invention may include, but are not limited to, glucose, galactose, mannose, fucose, n-acetylglucosamine and sialic acid.
  • the glycosylated binding protein comprises glycosyl residues such that the glycosylation pattern is human.
  • Differing protein glycosylation may result in differing protein characteristics.
  • the efficacy of a therapeutic protein produced in a microorganism host such as yeast, and
  • glycosylated utilizing the yeast endogenous pathway may be reduced compared to that of the same protein expressed in a mammalian cell, such as a CHO cell line.
  • a mammalian cell such as a CHO cell line.
  • Such glycoproteins may also be immunogenic in humans and show reduced half-life in vivo after administration.
  • Specific receptors in humans and other animals may recognize specific glycosyl residues and promote the rapid clearance of the protein from the bloodstream.
  • Other adverse effects may include changes in protein folding, solubility, susceptibility to proteases, trafficking, transport, compartmentalization, secretion, recognition by other proteins or factors, antigenicity, or allergenicity.
  • a practitioner may prefer a therapeutic protein with a specific composition and pattern of glycosylation, for example glycosylation composition and pattern identical, or at least similar, to that produced in human cells or in the species-specific cells of the intended subject animal
  • Expressing glycosylated proteins different from that of a host cell may be achieved by genetically modifying the host cell to express heterologous glycosylation enzymes.
  • a practitioner may generate antibodies or antigen binding portions thereof exhibiting human protein glycosylation.
  • yeast strains have been genetically modified to express non-naturally occurring glycosylation enzymes such that glycosylated proteins
  • yeast strains exhibit protein glycosylation identical to that of animal cells, especially human cells (U.S. patent Publication Nos.20040018590 and 20020137134 and PCT publication WO2005100584 A2).
  • Antibodies may be produced by any of a number of techniques. For example, expression from host cells, wherein expression vector(s) encoding the heavy and light chains is (are) transfected into a host cell by standard techniques.
  • the various forms of the term“transfection” are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE- dextran transfection and the like.
  • Preferred mammalian host cells for expressing the recombinant antibodies of the invention include Chinese Hamster Ovary (CHO cells) (including dhfr- CHO cells, described in Urlaub and Chasin, (1980) Proc. Natl. Acad. Sci. USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in R.J. Kaufman and P.A. Sharp (1982) Mol. Biol.159:601-621), NS0 myeloma cells, COS cells and SP2 cells.
  • Chinese Hamster Ovary CHO cells
  • dhfr- CHO cells described in Urlaub and Chasin, (1980) Proc. Natl. Acad. Sci. USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in R.J. Kaufman and P.A. Sharp (1982) Mol. Biol.159:601-621
  • NS0 myeloma cells
  • the antibodies When recombinant expression vectors encoding antibody genes are introduced into mammalian host cells, the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or, more preferably, secretion of the antibody into the culture medium in which the host cells are grown. Antibodies can be recovered from the culture medium using standard protein purification methods.
  • Host cells can also be used to produce functional antibody fragments, such as Fab fragments or scFv molecules. It will be understood that variations on the above procedure are within the scope of the invention. For example, it may be desirable to transfect a host cell with DNA encoding functional fragments of either the light chain and/or the heavy chain of an antibody of this invention. Recombinant DNA technology may also be used to remove some, or all, of the DNA encoding either or both of the light and heavy chains that is not necessary for binding to the antigens of interest. The molecules expressed from such truncated DNA molecules are also encompassed by the antibodies of the invention.
  • bifunctional antibodies may be produced in which one heavy and one light chain are an antibody of the invention and the other heavy and light chain are specific for an antigen other than the antigens of interest by crosslinking an antibody of the invention to a second antibody by standard chemical crosslinking methods.
  • a recombinant expression vector encoding both the antibody heavy chain and the antibody light chain is introduced into dhfr- CHO cells by calcium phosphate-mediated transfection.
  • the antibody heavy and light chain genes are each operatively linked to CMV enhancer/AdMLP promoter regulatory elements to drive high levels of transcription of the genes.
  • the recombinant expression vector also carries a DHFR gene, which allows for selection of CHO cells that have been transfected with the vector using methotrexate selection/amplification. The selected transformant host cells are cultured to allow for expression of the antibody heavy and light chains and intact antibody is recovered from the culture medium.
  • the invention provides a method of synthesizing a recombinant antibody of the invention by culturing a host cell in a suitable culture medium until a recombinant antibody is synthesized.
  • Recombinant antibodies of the invention may be produced using nucleic acid molecules corresponding to the amino acid sequences disclosed herein The method can further comprise isolating the recombinant antibody from the culture medium.
  • the N- and C-termini of antibody polypeptide chains of the present invention may differ from the expected sequence due to commonly observed post-translational modifications. For example, C- terminal lysine residues are often missing from antibody heavy chains. Dick et al. (2008) Biotechnol. Bioeng.100:1132. N-terminal glutamine residues, and to a lesser extent glutamate residues, are frequently converted to pyroglutamate residues on both light and heavy chains of therapeutic antibodies. Dick et al. (2007) Biotechnol. Bioeng.97:544; Liu et al. (2011) JBC 28611211; Liu et al. (2011) J. Biol. Chem.286:11211. III. Anti-B7-H3 Antibody Drug Conjugates (ADCs)
  • Anti-B7-H3 antibodies described herein may be conjugated to a drug moiety to form an anti- B7-H3 Antibody Drug Conjugate (ADC).
  • ADCs Antibody-drug conjugates
  • ADCs may increase the therapeutic efficacy of antibodies in treating disease, e.g., cancer, due to the ability of the ADC to selectively deliver one or more drug moiety(s) to target tissues, such as a tumor-associated antigen, e.g., B7-H3 expressing tumors.
  • the invention provides anti-B7-H3 ADCs for therapeutic use, e.g., treatment of cancer.
  • Anti-B7-H3 ADCs of the invention comprise an anti-B7-H3 antibody, i.e., an antibody that specifically binds to B7-H3, linked to one or more drug moieties.
  • the specificity of the ADC is defined by the specificity of the antibody, i.e., anti-B7-H3.
  • an anti-B7-H3 antibody is linked to one or more cytotoxic drug(s) which is delivered internally to a transformed cancer cell expressing B7-H3.
  • drugs that may be used in the anti-B7-H3 ADC of the invention are provided below, as are linkers that may be used to conjugate the antibody and the one or more drug(s).
  • linkers that may be used to conjugate the antibody and the one or more drug(s).
  • the terms“drug,”“agent,” and“drug moiety” are used interchangeably herein.
  • the terms“linked” and “conjugated” are also used interchangeably herein and indicate that the antibody and moiety are covalently linked.
  • the ADC has the following formula (formula I): wherein Ab is the antibody, e.g., anti-B7-H3 antibody huAb13v1, huAb3v2.5, or huAb3v2.6, and (L) is a linker, (D) is a drug, and LK represents a covalent linkage linking linker L to antibody Ab; and m is an integer ranging from 1 to 20.
  • D is a drug moiety having, for example, cytostatic, cytotoxic, or otherwise therapeutic activity against a target cell, e.g., a cell expressing B7-H3.
  • Ab is the antibody, e.g., anti-B7-H3 antibody huAb13v1, huAb3v2.5, or huAb3v2.6
  • L is a linker
  • D is a drug
  • LK represents a covalent linkage linking linker L to antibody Ab
  • m is an integer ranging from 1 to 20.
  • D is a drug
  • m ranges from 1 to 8, 1 to 7, 1 to 6, 2 to 6, 1 to 5, 1 to 4, 1 to 3, 1 to 2, 1.5 to 8, 1.5 to 7, 1.5 to 6, 1.5 to 5, 1.5 to 4, 2 to 6, 1 to 5, 1 to 4, 1 to 3, 1 to 2,or 2 to 4.
  • the DAR of an ADC is equivalent to the“m” referred to in Formula I.
  • the ADC has a formula of Ab- (LK-L-D) m , wherein Ab is an anti-B7-H3 antibody, e.g.huAb13v1, huAb3v2.5, or huAb3v2.6, L is a linker, D is a drug, e.g., a Bcl-xL inhibitor, LK is a covalent linker, e.g., -S-, and m is 1 to 8 (or a DAR of 2-4). Additional details regarding drugs (D of Formula I) and linkers (L of Formula I) that may be used in the ADCs of the invention, as well as alternative ADC structures, are described below. III. A.
  • Anti-B7-H3 ADCs Bcl-xL Inhibitors, Linkers, Synthons, and Methods of Making Same Dysregulated apoptotic pathways have also been implicated in the pathology of cancer.
  • the implication that down-regulated apoptosis (and more particularly the Bcl-2 family of proteins) is involved in the onset of cancerous malignancy has revealed a novel way of targeting this still elusive disease.
  • Research has shown, for example, the anti-apoptotic proteins, Bcl 2 and Bcl-xL, are over- expressed in many cancer cell types. See, Zhang, 2002, Nature Reviews/Drug Discovery 1:101;
  • aspects of the disclosure concern anti-B7-H3 ADCs comprising an anti-B7-H3 antibody conjugated to a drug via a linker, wherein the drug is a Bcl-xL inhibitor.
  • the ADCs are compounds according to structural formula (I) below, or a pharmaceutically acceptable salt thereof, wherein Ab represents the anti-B7-H3 antibody, D represents a Bcl-xL inhibitor drug (i.e., a compound of formula IIa or IIb as shown below), L represents a linker, LK represents a covalent linkage linking the linker (L) to the anti-B7-H3 antibody (Ab) and m represents the number of D-L- LK units linked to the antibody, which is an integer ranging from 1 to 20.
  • m is 2, 3 or 4. In some embodiments, m ranges from 1 to 8, 1 to 7, 1 to 6, 2 to 6, 1 to 5, 1 to 4, 1 to 3, 1 to 2, or 2 to 4. Specific embodiments of various Bcl-xL inhibitors per se, and various Bcl-xL inhibitors (D), linkers (L) and anti-B7-H3 antibodies (Ab) that can comprise the ADCs described herein, as well as the number of Bcl-xL inhibitors linked to the ADCs, are described in more detail below.
  • Bcl-xL inhibitors that may be used in the anti-B7-H3 ADC of the invention are provided below, as are linkers that may be used to conjugate the antibody and the one or more Bcl-xL inhibitor(s).
  • linkers that may be used to conjugate the antibody and the one or more Bcl-xL inhibitor(s).
  • the terms“linked” and“conjugated” are also used interchangeably herein and indicate that the antibody and moiety are covalently linked.
  • the Bcl-xL inhibitors may be used as compounds or salts per se in the various methods described herein, or may be included as a component part of an ADC, e.g., as the drug (D) in formula (I).
  • Bcl-xL inhibitors that may be used in unconjugated form, or that may be included as part of an ADC include compounds according to structural formula (IIa) or (IIb).
  • # shown in formula (IIa) or (IIb) below represents a point of attachment to a linker, which indicates that they are represented in a monoradical form.
  • Z 1 is selected from N, CH, C-halo and C-CN;
  • Z 2a , Z 2b , and Z 2c are each, independent from one another, selected from a bond, NR 6 , CR 6a R 6b , O, S, S(O), S(O) 2 , NR 6 C(O), NR 6a C(O)NR 6b , and NR 6 C(O)O;
  • R 1 is selected from hydrogen, methyl, halo, halomethyl, ethyl and cyano;
  • R 2 is selected from hydrogen, methyl, halo, halomethyl and cyano
  • R 3 is selected from hydrogen, lower alkyl and lower heteroalkyl
  • R 4 is selected from hydrogen, lower alkyl, monocyclic cycloalkyl, monocyclic heterocyclyl, lower heteroalkyl or is taken together with an atom of R 13 to form a cycloalkyl or heterocyclyl ring having between 3 and 7 ring atoms, wherein the lower alkyl, monocyclic cycloalkyl, monocyclic heterocyclyl, lower heteroalkyl are optionally substituted with one or more halo, cyano, C 1-4 alkoxy, monocyclic cycloalkyl, monocyclic heterocyclyl, NHC(O)CR 6a R 6b , NHS(O)CR 6a R 6b ,
  • R 6 , R 6a and R 6b are each, independent from one another, selected from hydrogen, lower alkyl, lower heteroalkyl, optionally substituted monocyclic cycloalklyl and monocyclic heterocyclyl, or are taken together with an atom from R 13 to form a cycloalkyl or heterocyclyl ring having between 3 and 7 ring atoms;
  • R 10 is selected from cyano, OR 14 , SR 14 , SOR 14 , SO 2 R 14 , SO 2 NR 14a R 14b , NR 14a R 14b , NHC(O)R 14 and NHSO 2 R 14 ;
  • R 11a and R 11b are each, independently of one another, selected from hydrogen, halo, methyl, ethyl, halomethyl, hydroxyl, methoxy, CN, and SCH 3 ;
  • R 12 is selected from hydrogen, halo, cyano, lower alkyl, lower heteroalkyl, cycloalkyl, or heterocyclyl, wherein the alkyl, heteroalkyl, cycloalkyl, or heterocyclyl are optionally substituted with one or more halo, cyano, C 1-4 alkoxy, monocyclic cycloalkyl, monocyclic heterocyclyl,
  • R 13 is selected from a bond, optionally substituted lower alkylene, optionally substituted lower heteroalkylene, optionally substituted cycloalkyl or optionally substituted heterocyclyl;
  • R 14 is selected from hydrogen, optionally substituted lower alkyl and optionally substituted lower heteroalkyl
  • R 14a and R 14b are each, independently of one another, selected from hydrogen, optionally substituted lower alkyl, optionally substituted lower heteroalkyl, or are taken together with the nitrogen atom to which they are bonded to form a monocyclic cycloalkyl or monocyclic heterocyclyl ring;
  • R 15 is selected from hydrogen, halo, C 1-6 alkanyl, C 2-4 alkenyl, C 2-4 alkynyl, and C 1-4 haloalkyl and C 1-4 hydroxyalkyl, with the proviso that when R 15 is present, R 4 is not C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, C 1-4 haloalkyl or C 1-4 hydroxyalkyl, wherein the R 4 C 1-6 alkanyl, C 2-4 alkenyl, C 2-4 alkynyl, C 1- 4 haloalkyl and C 1-4 hydroxyalkyl are optionally substituted with one or more substituents
  • # represents a point of attachment to a linker or a hydrogen atom.
  • Bcl-xL inhibitors that may be used in unconjugated form, or that may be included as part of an ADC include compounds according to structural formula (IIa) or (IIb): (IIa)
  • Z 1 is selected from N, CH, C-halo and C-CN;
  • Z 2a , Z 2b , and Z 2c are each, independent from one another, selected from a bond, NR 6 , CR 6a R 6b , O, S, S(O), S(O) 2 , NR 6 C(O), NR 6a C(O)NR 6b , and NR 6 C(O)O;
  • R 1 is selected from hydrogen, methyl, halo, halomethyl, ethyl and cyano;
  • R 2 is selected from hydrogen, methyl, halo, halomethyl and cyano
  • R 3 is selected from hydrogen, lower alkyl and lower heteroalkyl
  • R 4 is selected from hydrogen, lower alkyl, monocyclic cycloalkyl, monocyclic heterocyclyl, and lower heteroalkyl or is taken together with an atom of R 13 to form a cycloalkyl or heterocyclyl ring having between 3 and 7 ring atoms, wherein the lower alkyl, monocyclic cycloalkyl, monocyclic heterocyclyl, and lower heteroalkyl are optionally substituted with one or more halo, cyano, hydroxy, C 1-4 alkoxy, monocyclic cycloalkyl, monocyclic heterocyclyl, C(O)NR 6a R 6b , S(O) 2 NR 6a R 6b ,
  • R 6 , R 6a and R 6b are each, independent from one another, selected from hydrogen, lower alkyl, lower heteroalkyl, optionally substituted monocyclic cycloalklyl and monocyclic heterocyclyl, or are taken together with an atom from R 13 to form a cycloalkyl or heterocyclyl ring having between 3 and 7 ring atoms;
  • R 10 is selected from cyano, OR 14 , SR 14 , SOR 14 , SO 2 R 14 , SO 2 NR 14a R 14b , NR 14a R 14b , NHC(O)R 14 and NHSO 2 R 14 ;
  • R 11a and R 11b are each, independently of one another, selected from hydrogen, halo, methyl, ethyl, halomethyl, hydroxyl, methoxy, CN, and SCH 3 ;
  • R 12 is selected from hydrogen, halo, cyano, lower alkyl, lower heteroalkyl, cycloalkyl, and heterocyclyl, wherein the alkyl, heteroalkyl, cycloalkyl, and heterocyclyl are optionally substituted with one or more halo, cyano, C 1-4 alkoxy, monocyclic cycloalkyl, monocyclic heterocyclyl,
  • R 13 is selected from a bond, optionally substituted lower alkylene, optionally substituted lower heteroalkylene, optionally substituted cycloalkyl or optionally substituted heterocyclyl;
  • R 14 is selected from hydrogen, optionally substituted lower alkyl and optionally substituted lower heteroalkyl;
  • R 14a and R 14b are each, independently of one another, selected from hydrogen, optionally substituted lower alkyl, and optionally substituted lower heteroalkyl, or are taken together with the nitrogen atom to which they are bonded to form an optionally substituted monocyclic cycloalkyl or monocyclic heterocyclyl ring;
  • R 15 is selected from hydrogen, halo, C 1-6 alkanyl, C 2-4 alkenyl, C 2-4 alkynyl, and C 1-4 haloalkyl and C 1-4 hydroxyalkyl, with the proviso that when R 15 is present, R 4 is not C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, C 1-4 haloalkyl or C 1-4 hydroxyalkyl, wherein the R 4 C 1-6 alkanyl, C 2-4 alkenyl, C 2-4 alkynyl, C 1- 4 haloalkyl and C 1-4 hydroxyalkyl are optionally substituted with one or more substituents
  • # represents a point of attachment to a linker or a hydrogen atom.
  • Bcl-xL inhibitors that may be used in unconjugated form, or that may be included as part of an ADC include compounds according to structural formula (IIa) or (IIb):
  • Ar 1 is selected from ,
  • Z 1 is selected from N, CH, C-halo and C-CN;
  • Z 2a , Z 2b , and Z 2c are each, independent from one another, selected from a bond, NR 6 , CR 6a R 6b , O, S, S(O), SO 2 , NR 6 C(O), NR 6a C(O)NR 6b , and NR 6 C(O)O;
  • R 1 is selected from hydrogen, methyl, halo, halomethyl, ethyl and cyano;
  • R 2 is selected from hydrogen, methyl, halo, halomethyl and cyano
  • R 3 is selected from hydrogen, lower alkyl and lower heteroalkyl
  • R 4 is selected from hydrogen, lower alkyl, monocyclic cycloalkyl, monocyclic heterocyclyl, lower heteroalkyl or is taken together with an atom of R 13 to form a cycloalkyl or heterocyclyl ring having between 3 and 7 ring atoms, wherein the lower alkyl, monocyclic cycloalkyl, monocyclic heterocyclyl, lower heteroalkyl are optionally substituted with one or more halo, cyano, C 1-4 alkoxy, monocyclic cycloalkyl, monocyclic heterocyclyl, NC(O)CR 6a R 6b , NS(O)CR 6a R 6b , NS(O) 6a
  • R 6 , R 6a and R 6b are each, independent from one another, selected from hydrogen, lower alkyl, lower heteroalkyl, optionally substituted monocyclic cycloalklyl and monocyclic heterocyclyl, or are taken together with an atom from R 13 to form a cycloalkyl or heterocyclyl ring having between 3 and 7 ring atoms;
  • R 10 is selected from cyano, OR 14 , SR 14 , SOR 14 , SO 2 R 14 , SO 2 NR 14a R 14b , NR 14a R 14b , NHC(O)R 14 and NHSO 2 R 14 ;
  • R 11a and R 11b are each, independently of one another, selected from hydrogen, halo, methyl, ethyl, halomethyl, hydroxyl, methoxy, CN, and SCH 3 ;
  • R 12 is selected from hydrogen, halo, cyano, lower alkyl, lower heteroalkyl, cycloalkyl, or heterocyclyl, wherein the alkyl, heteroalkyl, cycloalkyl, or heterocyclyl are optionally substituted with one or more halo, cyano, C 1-4 alkoxy, monocyclic cycloalkyl, monocyclic heterocyclyl,
  • R 13 is selected from a bond, optionally substituted lower alkylene, optionally substituted lower heteroalkylene, optionally substituted cycloalkyl or optionally substituted heterocyclyl;
  • R 14 is selected from hydrogen, optionally substituted lower alkyl and optionally substituted lower heteroalkyl
  • R 14a and R 14b are each, independently of one another, selected from hydrogen, optionally substituted lower alkyl, optionally substituted lower heteroalkyl, or are taken together with the nitrogen atom to which they are bonded to form a monocyclic cycloalkyl or monocyclic heterocyclyl ring;
  • R 15 is selected from hydrogen, halo, C 1-6 alkanyl, C 2-4 alkenyl, C 2-4 alkynyl, and C 1-4 haloalkyl and C 1-4 hydroxyalkyl, with the proviso that when R 15 is present, R 4 is not C 1-4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, C 1-4 haloalkyl or C 1-4 hydroxyalkyl, wherein the R 4 C 1-6 alkanyl, C 2-4 alkenyl, C 2-4 alkynyl, C 1- 4 haloalkyl and C 1-4 hydroxyalkyl are optionally substituted with one or more substituents
  • # represents a point of attachment to a linker or a hydrogen atom.
  • Ar 1 is .
  • Ar 1 is unsubstituted.
  • the #-N(R 4 )-R 13 -Z 2b - substituent of formula (IIb) is attached to Ar 2 at any Ar 2 atom capable of being substituted.
  • Ar 2 of formula (IIa) or (IIb) is which is optionally substituted at the 5- osition with a rou selected from h drox l C 1-4 alkox and cyano; or
  • Ar 2 of formula (IIa) or (IIb) is In certain embodiments, Ar 2 of formula (IIa) or (IIb) is In certain embodiments, Ar 2 of formula (IIa) or (IIb) is . In certain embodiments, Ar 2 of formula (IIa) or (IIb) is In certain embodiments, Ar 2 of formula (IIa) or (IIb) is
  • Ar 2 of formula (IIa) or (IIb) is
  • Ar 2 of formula (IIa) or (IIb) is In certain embodiments, Ar 2 of formula (IIa) or (IIb) is In certain embodiments, Ar 2 of formula (IIa) or (IIb) is
  • Ar 2 of formula (IIa) or (IIb) is In certain embodiments, Ar 2 of formula (IIa) or (IIb) is In certain embodiments, Ar 2 of formula (IIa) or (IIb) is
  • Ar 2 of formula (IIa) or (IIb) is
  • Ar 2 of formula (IIa) or (IIb) is In certain embodiments, Ar 2 of formula (IIa) or (IIb) is In certain embodiments, Ar 2 of formula (IIa) or (IIb) is . In certain embodiments, Ar 2 of formula (IIa) or (IIb) is
  • Ar 2 of formula (IIa) or (IIb) is In certain embodiments, Ar 2 of formula (IIa) or (IIb) is In certain embodiments, Ar 2 of formula (IIa) or (IIb) is In certain embodiments, Ar 2 of formula (IIa) or (IIb) is In certain embodiments, Ar 2 of formula (IIa) or (IIb) is In certain embodiments, Ar 2 of formula (IIa) or (IIb) is In certain embodiments, Ar 2 of formula (IIa) or (IIb) is
  • Ar 2 of formula (IIa) or (IIb) is . In certain embodiments, Ar 2 of formula (IIa) is unsubstituted.
  • Ar 2 of formula (IIa) or (IIb) is , which is substituted at the 5-position with a group selected from hydroxyl, C 1-4 alkoxy, and cyano.
  • Z 1 of formula (IIa) or (IIb) is N.
  • R 1 of formula (IIa) or (IIb) is selected from methyl and chloro.
  • R 2 of formula (IIa) or (IIb) is selected from hydrogen and methyl. In particular embodiments, R 2 is hydrogen.
  • R 4 of formula (IIa) or (IIb) is methyl.
  • R 4 of formula (IIa) or (IIb) is (CH 2 ) 2 OCH 3 .
  • R 4 of formula (IIa) or (IIb) is hydrogen.
  • R 4 of formula (IIa) or (IIb) is monocyclic heterocyclyl, wherein the monocyclic heterocycloalkyl is substituted with one S(O) 2 CH 3 .
  • R 4 of formula (IIa) or (IIb) is hydrogen or lower alkyl, wherein the lower alkyl is optionally substituted with C 1-4 alkoxy or C(O)NR 6a R 6b .
  • R 4 of formula (IIa) or (IIb) is lower alkyl, wherein the lower alkyl is substituted with C(O)NH 2 .
  • R 4 of formula (IIa) or (IIb) is lower alkyl, wherein the lower alkyl is substituted with S(O) 2 NH 2 .
  • R 4 of formula (IIa) or (IIb) is lower alkyl, wherein the lower alkyl is substituted with hydroxy.
  • R 4 of formula (IIa) or (IIb) is lower alkyl, wherein the lower alkyl is substituted with C(O)N(CH 3 ) 2 .
  • R 4 of formula (IIa) or (IIb) is lower alkyl, wherein the lower alkyl is substituted with C(O)NHCH 3 .
  • R 11a and R 11b of formula (IIa) or (IIb) are the same. In a particular embodiment, R 11a and R 11b are each methyl. In another embodiment, R 11a and R 11b are each ethyl. In another embodiment, R 11a and R 11b are each methoxy. In certain embodiments, R 11a and R 11b of formula (IIa) or (IIb) are independently selected from F, Br and Cl.
  • Z 1 is N
  • Z 2a is O
  • R 1 is methyl or chloro
  • R 2 is hydrogen
  • Ar 2 is is optionally substituted at the 5-position with a group selected from hydroxyl, C 1-4 alkoxy, and cyano.
  • Certain embodiments pertain to a compound of formula (IIa).
  • Z 2a of formula (IIa) is O.
  • Z 2a of formula (IIa) is CH 2 or O.
  • Z 2a of formula (IIa) is S.
  • Z 2a of formula (IIa) is CH 2 .
  • Z 2a of formula (IIa) is NR 6 . In some such embodiments R 6 is methyl. In certain embodiments, Z 2a of formula (IIa) is NR 6 C(O). In some such embodiments R 6 is hydrogen.
  • Z 2a of formula (IIa) is O, R 13 is ethylene, and R 4 is lower alkyl. In certain embodiments, Z 2a of formula (IIa) is O, R 13 is ethylene, and R 4 is hydrogen or lower alkyl optionally substituted with C 1-4 alkoxy or C(O)NR 6a R 6b .
  • Z 2a of formula (IIa) is O, R 13 is ethylene, and R 4 is methyl.
  • Z 2a of formula (IIa) is O, R 13 is ethylene, and R 4 is hydrogen.
  • Z 2a of formula (IIa) is NR 6 C(O), R 6 is hydrogen, R 13 is methylene, and R 4 is hydrogen.
  • Z 2a of formula (IIa) is S, R 13 is ethylene, and R 4 is hydrogen.
  • Z 2a of formula (IIa) is CH 2 , R 13 is ethylene, and R 4 is hydrogen. In certain embodiments, the group R 13 in formula (IIa) is ethylene. In some such
  • embodiments Z 2a is O.
  • the group R 13 in formula (IIa) is propylene. In some such embodiments Z 2a is O.
  • the group R 13 in formula (IIa) is selected from lower alkylene or lower heteroalkylene.
  • the group R 13 in formula (IIa) is selected from (CH 2 ) 2 O(CH 2 ) 2 , (CH 2 ) 3 O(CH 2 ) 2 , (CH 2 ) 2 O(CH 2 ) 3 and (CH 2 ) 3 O(CH 2 ) 3 .
  • Z 2a is O.
  • the group R 13 in formula (IIa) is selected from (CH 2 ) 2 (SO 2 )(CH 2 ) 2 , (CH 2 ) 3 (SO 2 )(CH 2 ) 2 , (CH 2 ) 2 (SO 2 )(CH 2 ) 3 and (CH 2 ) 3 (SO 2 )(CH 2 ) 3 .
  • Z 2a is O.
  • the group R 13 in formula (IIa) is selected from (CH 2 ) 2 (SO)(CH 2 ) 2 , (CH 2 ) 2 (SO)(CH 2 ) 3 , (CH 2 ) 3 (SO)(CH 2 ) 2 and (CH 2 ) 3 (SO)(CH 2 ) 3 .
  • Z 2a is O.
  • the group R 13 in formula (IIa) is selected from (CH 2 ) 2 S(CH 2 ) 2 , (CH 2 ) 2 S(CH 2 ) 3 , (CH 2 ) 3 S(CH 2 ) 2 and (CH 2 ) 3 S(CH 2 ) 3 .
  • Z 2a is O.
  • the group Z 2b in formula (IIb) is a bond, O, or NR 6 , or and R 13 is ethylene or optionally substituted heterocyclyl.
  • the group Z 2b in formula (IIb) is NR 6 .
  • R 6 is methyl.
  • the group Z 2b in formula (IIb) is NR 6 and R 13 is ethylene. In some such embodiments R 6 is methyl.
  • the group Z 2b in formula (IIb) is O and R 13 is ethylene. In some such embodiments R 4 is methyl.
  • the group Z 2b in formula (IIb) is NR 6 , wherein the R 6 group is taken together with an atom of R 13 to form a ring having between 4 and 6 atoms.
  • the ring is a five membered ring.
  • the group Z 2b in formula (IIb) is methylene and the group R 13 is methylene.
  • the group Z 2b in formula (IIb) is methylene and the group R 13 is a bond.
  • the group Z 2b in formula (IIb) is oxygen and the group R 13 is selected from (CH 2 ) 2 O(CH 2 ) 2 , (CH 2 ) 3 O(CH 2 ) 2 , (CH 2 ) 2 O(CH 2 ) 3 and (CH 2 ) 3 O(CH 2 ) 3 .
  • the group R 13 is selected from (CH 2 ) 2 O(CH 2 ) 2 , (CH 2 ) 3 O(CH 2 ) 2 , (CH 2 ) 2 O(CH 2 ) 3 and (CH 2 ) 3 O(CH 2 ) 3 .
  • R 4 is methyl
  • the group Z 2c in formula (IIb) is a bond and R 12 is OH.
  • the group Z 2c in formula (IIb) is a bond and R 12 is selected from F, Cl, Br and I.
  • the group Z 2c in formula (IIb) is a bond and R 12 is lower alkyl. In some such embodiments R 12 is methyl.
  • the group Z 2c in formula (IIb) is O and R 12 is a lower heteroalkyl.
  • R 12 is O(CH 2 ) 2 OCH 3 .
  • the group Z 2c in formula (IIb) is O and R 12 is lower alkyl optionally substituted with one or more halo or C 1-4 alkoxy.
  • the group Z 2c in formula (IIb) is O and R 12 is a lower alkyl.
  • R 12 is methyl.
  • the group Z 2c in formula (IIb) is S and R 12 is a lower alkyl. In some such embodiments R 12 is methyl.
  • Exemplary Bcl-xL inhibitors according to structural formulae (IIa)-(IIb) that may be used in the methods described herein in unconjugated form and/or included in the ADCs described herein include the following compounds, and/or a pharmaceutically acceptable salt thereof:
  • the hydrogen corresponding to the # position of structural formula (IIa) or (IIb) is not present, forming a monoradical.
  • compound W3.01 (Example 1.1) is 6-[1-(1,3-benzothiazol-2- ylcarbamoyl)-1,2,3,4-tetrahydroquinolin-7-yl]-3-[1-( ⁇ 3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.1 3,7 ]dec-1-yl ⁇ methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid.
  • it is in uncon u ated form it has the followin structure:
  • the Bcl-xL inhibitor is selected from the group consisting of W3.01, W3.02, W3.03, W3.04, W3.05, W3.06, W3.07, W3.08, W3.09, W3.10, W3.11, W3.12, W3.13, W3.14, W3.15, W3.16, W3.17, W3.18, W3.19, W3.20, W3.21, W3.22, W3.23, W3.24, W3.25, W3.26, W3.27, W3.28, W3.29, W3.30, W3.31, W3.32, W3.33, W3.34, W3.35, W3.36, W3.37, W3.38, W3.39, W3.40, W3.41, W3.42, W3.43, and pharmaceutically acceptable salts thereof (see Example 1 for compounds).
  • the ADC comprises a drug linked to an antibody by way of a linker, wherein the drug is a Bcl-xL inhibitor selected from the group consisting of W3.01, W3.02, W3.03, W3.04, W3.05, W3.06, W3.07, W3.08, W3.09, W3.10, W3.11, W3.12, W3.13, W3.14, W3.15, W3.16, W3.17, W3.18, W3.19, W3.20, W3.21, W3.22, W3.23, W3.24, W3.25, W3.26, W3.27, W3.28, W3.29, W3.30, W3.31, W3.32, W3.33, W3.34, W3.35, W3.36, W3.37, W3.38, W3.39, W3.40, W3.41, W3.42, W3.43.
  • the drug is a Bcl-xL inhibitor selected from the group consisting of W3.01, W3.02, W3.03, W3.04, W3.05, W3.06, W3.07, W3.08, W3.09, W3.10, W3.11, W3.1
  • the ADC, or a pharmaceutically acceptable salt thereof, the Bcl-xL inhibitor is selected from the group consisting of the following compounds modified in that the hydrogen corresponding to the # position of structural formula (IIa) or (IIb) is not present forming a monoradical:
  • the Bcl-xL inhibitors bind to and inhibit anti-apoptotic Bcl-xL proteins, inducing apoptosis.
  • the ability of specific Bcl-xL inhibitors according to structural formulae (IIa)-(IIb) to bind to and inhibit Bcl-xL activity may be confirmed in standard binding and activity assays, including, for example, the TR-FRET Bcl-xL binding assays described in Tao et al., 2014, ACS Med. Chem. Lett., 5:1088-1093.
  • a specific TR-FRET Bcl-xL binding assay that can be used to confirm Bcl-xL binding is provided in Example 4, below.
  • Bcl-xL inhibitors useful as inhibitors per se and in the ADCs described herein will exhibit a K i in the binding assay of Example 5 of less than about 1 nM, but may exhibit a significantly lower K i , for example a K i of less than about 1, 0.1, or even 0.01.
  • Bcl-xL inhibitory activity may also be confirmed in standard cell-based cytotoxicity assays, such as the FL5.12 cellular and Molt-4 cytotoxicity assays described in Tao et al., 2014, ACS Med. Chem. Lett., 5:1088-1093.
  • standard cell-based cytotoxicity assays such as the FL5.12 cellular and Molt-4 cytotoxicity assays described in Tao et al., 2014, ACS Med. Chem. Lett., 5:1088-1093.
  • a specific Molt-4 cellular cytotoxicity assay that may be used to confirm Bcl-xL inhibitory activity of specific Bcl-xL inhibitors that are able to permeate cell membranes is provided in Example 5, below.
  • such cell-permeable Bcl-xL inhibitors will exhibit an EC 50 of less than about 500 nM in the Molt-4 cytotoxicity assay of Example 5, but may exhibit a significantly lower EC 50 , for example an EC 50 of less than about 250, 100, 50, 20, 10 or even 5 nM.
  • MOMP mitochondrial outer-membrane permeabilization
  • Bcl-2 family proteins The process of mitochondrial outer-membrane permeabilization (MOMP) is controlled by the Bcl-2 family proteins. Specifically, MOMP is promoted by the pro-apoptotic Bcl-2 family proteins Bax and Bak which, upon activation oligomerize on the outer mitochondrial membrane and form pores, leading to release of cytochrome c (cyt c). The release of cyt c triggers formulation of the apoptosome which, in turn, results in caspase activation and other events that commit the cell to undergo programmed cell death (see, Goldstein et al., 2005, Cell Death and Differentiation 12:453- 462).
  • the oligomerization action of Bax and Bak is antagonized by the anti-apoptotic Bcl-2 family members, including Bcl-2 and Bcl-xL.
  • Bcl-xL inhibitors in cells that depend upon Bcl-xL for survival, can cause activation of Bax and/or Bak, MOMP, release of cyt c and downstream events leading to apoptosis.
  • the process of cyt c release can be assessed via western blot of both mitochondrial and cytosolic fractions of cytochrome c in cells and used as a proxy measurement of apoptosis in cells.
  • the cells can be treated with an agent that causes selective pore formation in the plasma, but not mitochondrial, membrane.
  • the cholesterol/phospholipid ratio is much higher in the plasma membrane than the mitochondrial membrane.
  • This agent forms insoluble complexes with cholesterol leading to the segregation of cholesterol from its normal phospholipid binding sites. This action, in turn, leads to the formation of holes about 40-50 ⁇ wide in the lipid bilayer.
  • cytosolic components able to pass over digitonin-formed holes can be washed out, including the cytochrome C that was released from mitochondria to cytosol in the apoptotic cells (Campos, 2006, Cytometry A 69(6):515-523).
  • Bcl-xL inhibitors of structural formulae (IIa)-(IIb) selectively or specifically inhibit Bcl-xL over other anti-apoptotic Bcl-2 family proteins, selective and/or specific inhibition of Bcl-xL is not necessary.
  • the Bcl-xL inhibitors and ADCs comprising the compounds may also, in addition to inhibiting Bcl-xL, inhibit one or more other anti-apoptotic Bcl-2 family proteins, such as, for example, Bcl-2.
  • the Bcl-xL inhibitors and/or ADCs are selective and/or specific for Bcl-xL.
  • Bcl-xL inhibitor and/or ADC binds or inhibits Bcl-xL to a greater extent than Bcl-2 under equivalent assay conditions.
  • the Bcl-xL inhibitors and/or ADCs exhibit in the range of about 10-fold, 100-fold, or even greater specificity or selectivity for Bcl-xL than Bcl-2 in binding assays.
  • the Bcl-xL inhibitors are linked to the anti-B7-H3 antibody by way of linkers.
  • the linker linking a Bcl-xL inhibitor to the anti-B7-H3 antibody of an ADC may be short, long, hydrophobic, hydrophilic, flexible or rigid, or may be composed of segments that each independently has one or more of the above-mentioned properties such that the linker may include segments having different properties.
  • the linkers may be polyvalent such that they covalently link more than one Bcl-xL inhibitor to a single site on the antibody, or monovalent such that covalently they link a single Bcl-xL inhibitor to a single site on the antibody.
  • the linkers link the Bcl-xL inhibitors to the anti- B7-H3 antibody by forming a covalent linkage to the Bcl-xL inhibitor at one location and a covalent linkage to antibody at another.
  • the covalent linkages are formed by reaction between functional groups on the linker and functional groups on the inhibitors and antibody.
  • the expression“linker” is intended to include (i) unconjugated forms of the linker that include a functional group capable of covalently linking the linker to a Bcl-xL inhibitor and a functional group capable of covalently linking the linker to an anti-B7-H3 antibody; (ii) partially conjugated forms of the linker that include a functional group capable of covalently linking the linker to an anti-B7-H3 antibody and that is covalently linked to a Bcl-xL inhibitor, or vice versa; and (iii) fully conjugated forms of the linker that is covalently linked to both a Bcl-xL inhibitor and an anti-B7-H3 antibody.
  • moieties comprising the functional groups on the linker and covalent linkages formed between the linker and antibody are specifically illustrated as R x and LK, respectively.
  • One embodiment pertains to an ADC formed by contacting an antibody that binds a cell surface receptor or tumor associated antigen expressed on a tumor cell with a synthon described herein under conditions in which the synthon covalently links to the anti-B7-H3 antibody.
  • One embodiment pertains to a method of making an ADC formed by contacting a synthon described herein under conditions in which the synthon covalently links to the anti-B7-H3 antibody.
  • One embodiment pertains to a method of inhibiting Bcl-xL activity in a cell that expresses Bcl-xL, comprising contacting the cell with an ADC described herein that is capable of binding the cell, under conditions in which the ADC binds the cell.
  • the Fleximer® linker technology is based on incorporating drug molecules into a solubilizing poly-acetal backbone via a sequence of ester bonds
  • the methodology renders highly-loaded ADCs (DAR up to 20) whilst maintaining good physicochemical properties.
  • This methodology could be utilized with Bcl-xL inhibitors as shown in the Scheme below.
  • an aliphatic alcohol can be present or introduced into the Bcl-xL inhibitor.
  • the alcohol moiety is then conjugated to an alanine moiety, which is then synthetically incorporated into the Fleximer® linker. Liposomal processing of the ADC in vitro releases the parent alcohol–containing drug.
  • Exemplary monovalent linkers that may be used are described, for example, in Nolting, 2013, Antibody-Drug Conjugates, Methods in Molecular Biology 1045:71-100; Kitson et al., 2013, CROs/CMOs - Chemica Oggi– Chemistry Today 31(4): 30-36; Ducry et al., 2010, Bioconjugate Chem.21:5-13; Zhao et al., 2011, J. Med. Chem.54:3606-3623; U.S. Patent No.7,223,837; U.S. Patent No.8,568,728; U.S. Patent No.8,535,678; and WO2004010957, the content of each of which is incorporated herein by reference in their entireties.
  • the linker selected is cleavable in vitro and in vivo.
  • Cleavable linkers may include chemically or enzymatically unstable or degradable linkages.
  • Cleavable linkers generally rely on processes inside the cell to liberate the drug, such as reduction in the cytoplasm, exposure to acidic conditions in the lysosome, or cleavage by specific proteases or other enzymes within the cell.
  • Cleavable linkers generally incorporate one or more chemical bonds that are either chemically or enzymatically cleavable while the remainder of the linker is noncleavable.
  • a linker comprises a chemically labile group such as hydrazone and/or disulfide groups.
  • Linkers comprising chemically labile groups exploit differential properties between the plasma and some cytoplasmic compartments.
  • the intracellular conditions to facilitate drug release for hydrazone containing linkers are the acidic environment of endosomes and lysosomes, while the disulfide containing linkers are reduced in the cytosol, which contains high thiol concentrations, e.g., glutathione.
  • the plasma stability of a linker comprising a chemically labile group may be increased by introducing steric hindrance using substituents near the chemically labile group.
  • Acid-labile groups such as hydrazone, remain intact during systemic circulation in the blood’s neutral pH environment (pH 7.3-7.5) and undergo hydrolysis and release the drug once the ADC is internalized into mildly acidic endosomal (pH 5.0-6.5) and lysosomal (pH 4.5-5.0) compartments of the cell.
  • This pH dependent release mechanism has been associated with nonspecific release of the drug
  • the linker may be varied by chemical modification, e.g., substitution, allowing tuning to achieve more efficient release in the lysosome with a minimized loss in circulation.
  • Hydrazone-containing linkers may contain additional cleavage sites, such as additional acid- labile cleavage sites and/or enzymatically labile cleavage sites.
  • ADCs including exemplary hydrazone-containing linkers include the following structures:
  • linker (Ig) the linker comprises two cleavable groups– a disulfide and a hydrazone moiety.
  • linkers such as (Ih) and (Ii) have been shown to be effective with a single hydrazone cleavage site.
  • linkers include cis-aconityl-containing linkers.
  • cis-Aconityl chemistry uses a carboxylic acid juxtaposed to an amide bond to accelerate amide hydrolysis under acidic conditions.
  • Cleavable linkers may also include a disulfide group.
  • Disulfides are thermodynamically stable at physiological pH and are designed to release the drug upon internalization inside cells, wherein the cytosol provides a significantly more reducing environment compared to the extracellular environment. Scission of disulfide bonds generally requires the presence of a cytoplasmic thiol cofactor, such as (reduced) glutathione (GSH), such that disulfide-containing linkers are reasonable stable in circulation, selectively releasing the drug in the cytosol.
  • GSH cytoplasmic thiol cofactor
  • the intracellular enzyme protein disulfide isomerase or similar enzymes capable of cleaving disulfide bonds, may also contribute to the preferential cleavage of disulfide bonds inside cells.
  • GSH is reported to be present in cells in the concentration range of 0.5-10 mM compared with a significantly lower concentration of GSH or cysteine, the most abundant low-molecular weight thiol, in circulation at approximately 5 ⁇ M.
  • Tumor cells where irregular blood flow leads to a hypoxic state, result in enhanced activity of reductive enzymes and therefore even higher glutathione concentrations.
  • the in vivo stability of a disulfide-containing linker may be enhanced by chemical modification of the linker, e.g., use of steric hindrance adjacent to the disulfide bond.
  • ADCs including exemplary disulfide-containing linkers include the following structures:
  • n represents the number of drug- linkers linked to the anti-B7-H3 antibody and R is independently selected at each occurrence from hydrogen or alkyl, for example.
  • R is independently selected at each occurrence from hydrogen or alkyl, for example.
  • increasing steric hindrance adjacent to the disulfide bond increases the stability of the linker.
  • Structures such as (Ij) and (Il) show increased in vivo stability when one or more R groups is selected from a lower alkyl such as methyl.
  • linker that is specifically cleaved by an enzyme.
  • the linker is cleavable by a lysosomal enzyme.
  • Such linkers are typically peptide- based or include peptidic regions that act as substrates for enzymes.
  • Peptide based linkers tend to be more stable in plasma and extracellular milieu than chemically labile linkers. Peptide bonds generally have good serum stability, as lysosomal proteolytic enzymes have very low activity in blood due to endogenous inhibitors and the unfavorably high pH value of blood compared to lysosomes.
  • the linker is cleavable by a lysosomal enzyme.
  • the linker is cleavable by a lysosomal enzyme, and the lysosomal enzyme is Cathepsin B.
  • the linker is cleavable by a lysosomal enzyme, and the lysosomal enzyme is ⁇ -glucuronidase or ⁇ -galactosidase.
  • the linker is cleavable by a lysosomal enzyme, and the lysosomal enzyme is ⁇ -glucuronidase. In certain embodiments, the linker is cleavable by a lysosomal enzyme, and the lysosomal enzyme is ⁇ -galactosidase.
  • the cleavable peptide is selected from tetrapeptides such as Gly-Phe-Leu-Gly, Ala-Leu-Ala-Leu or dipeptides such as Val-Cit, Val-Ala, and Phe-Lys.
  • dipeptides are preferred over longer polypeptides due to hydrophobicity of the longer peptides.
  • dipeptide linkers that may be used include those found in ADCs such as Seattle Genetics’ Brentuximab Vendotin SGN-35 (AdcetrisTM), Seattle Genetics SGN-75 (anti-CD-70, MC- monomethyl auristatin F(MMAF), Celldex Therapeutics glembatumumab (CDX-011) (anti-NMB, Val-Cit- monomethyl auristatin E(MMAE), and Cytogen PSMA-ADC (PSMA-ADC-1301) (anti- PSMA, Val-Cit-MMAE).
  • ADCs such as Seattle Genetics’ Brentuximab Vendotin SGN-35 (AdcetrisTM), Seattle Genetics SGN-75 (anti-CD-70, MC- monomethyl auristatin F(MMAF), Celldex Therapeutics glembatumumab (CDX-011) (anti-NMB, Val-Cit- monomethyl auristatin E(MMAE), and Cytogen PS
  • Enzymatically cleavable linkers may include a self-immolative spacer to spatially separate the drug from the site of enzymatic cleavage.
  • the direct attachment of a drug to a peptide linker can result in proteolytic release of an amino acid adduct of the drug, thereby impairing its activity.
  • the use of a self-immolative spacer allows for the elimination of the fully active, chemically unmodified drug upon amide bond hydrolysis.
  • One self-immolative spacer is the bifunctional para-aminobenzyl alcohol group, which is linked to the peptide through the amino group, forming an amide bond, while amine containing drugs may be attached through carbamate functionalities to the benzylic hydroxyl group of the linker (to give a p-amidobenzylcarbamate, PABC).
  • the resulting prodrugs are activated upon protease-mediated cleavage, leading to a 1,6-elimination reaction releasing the unmodified drug, carbon dioxide, and remnants of the linker group.
  • the following scheme depicts the fragmentation of p-amidobenzyl carbamate and release of the drug:
  • the enzymatically cleavable linker is a ß-glucuronic acid-based linker. Facile release of the drug may be realized through cleavage of the ß-glucuronide glycosidic bond by the lysosomal enzyme ß-glucuronidase. This enzyme is present abundantly within lysosomes and is overexpressed in some tumor types, while the enzyme activity outside cells is low.
  • ß- Glucuronic acid-based linkers may be used to circumvent the tendency of an ADC to undergo aggregation due to the hydrophilic nature of ß-glucuronides.
  • ß-glucuronic acid-based linkers are preferred as linkers for ADCs linked to hydrophobic drugs. The following scheme depicts the release of the drug from and ADC containing a ß-glucuronic acid-based linker:
  • cleavable ß-glucuronic acid-based linkers useful for linking drugs such as auristatins, camptothecin and doxorubicin analogues, CBI minor-groove binders, and psymberin to antibodies have been described (see, Jeffrey et al., 2006, Bioconjug. Chem.17:831-840; Jeffrey et al., 2007, Bioorg. Med. Chem. Lett.17:2278-2280; and Jiang et al., 2005, J. Am. Chem. Soc.127:11254- 11255, the contents of each of which are incorporated herein by reference).
  • the enzymatically cleavable linker is a ß-galactoside-based linker.
  • ß-Galactoside is present abundantly within lysosomes, while the enzyme activity outside cells is low.
  • Bc1-xL inhibitors containing a phenol group can be covalently bonded to a linker through the phenolic oxygen.
  • a linker described in U.S. Patent App. No.
  • Cleavable linkers may include noncleavable portions or segments, and/or cleavable segments or portions may be included in an otherwise non-cleavable linker to render it cleavable.
  • polyethylene glycol (PEG) and related polymers may include cleavable groups in the polymer backbone.
  • a polyethylene glycol or polymer linker may include one or more cleavable groups such as a disulfide, a hydrazone or a dipeptide.
  • degradable linkages that may be included in linkers include ester linkages formed by the reaction of PEG carboxylic acids or activated PEG carboxylic acids with alcohol groups on a biologically active agent, wherein such ester groups generally hydrolyze under physiological conditions to release the biologically active agent.
  • Hydrolytically degradable linkages include, but are not limited to, carbonate linkages; imine linkages resulting from reaction of an amine and an aldehyde; phosphate ester linkages formed by reacting an alcohol with a phosphate group; acetal linkages that are the reaction product of an aldehyde and an alcohol; orthoester linkages that are the reaction product of a formate and an alcohol; and oligonucleotide linkages formed by a
  • phosphoramidite group including but not limited to, at the end of a polymer, and a 5' hydroxyl group of an oligonucleotide.
  • the linker comprises an enzymatically cleavable peptide moiety, for example, a linker comprising structural formula (IVa), (IVb), (IVc), or (IVd):
  • peptide represents a peptide (illustrated N ⁇ C, wherein peptide includes the amino and carboxy“termini”) a cleavable by a lysosomal enzyme;
  • T represents a polymer comprising one or more ethylene glycol units or an alkylene chain, or combinations thereof;
  • R a is selected from hydrogen, C 1-6 alkyl, SO 3 H and CH 2 SO 3 H;
  • R y is hydrogen or C 1-4 alkyl-(O) r -(C 1-4 alkylene) s -G 1 or C 1-4 alkyl-(N)-[(C 1-4 alkylene)-G 1 ] 2 ;
  • R z is C 1-4 alkyl-(O) r -(C 1-4 alkylene) s -G 2 ;
  • G 1 is SO 3 H, CO 2 H, PEG 4-32, or sugar moiety
  • G 2 is SO 3 H, CO 2 H, or PEG 4-32 moiety
  • r is 0 or 1;
  • s is 0 or 1;
  • p is an integer ranging from 0 to 5;
  • q is 0 or 1
  • x is 0 or 1
  • y is 0 or 1
  • the linker comprises an enzymatically cleavable peptide moiety, for example, a linker comprising structural formula (IVa), (IVb), (IVc), (IVd) or a pharmaceutically acceptable salt thereof.
  • linker L comprises a segment according to structural formula IVa or IVb or a pharmaceutically acceptable salt thereof.
  • the peptide is selected from a tripeptide or a dipeptide.
  • the dipeptide is selected from: Val-Cit; Cit-Val; Ala-Ala; Ala-Cit; Cit-Ala; Asn-Cit; Cit-Asn; Cit-Cit; Val-Glu; Glu-Val; Ser-Cit; Cit-Ser; Lys-Cit; Cit-Lys; Asp-Cit; Cit-Asp; Ala-Val; Val-Ala; Phe-Lys; Lys-Phe; Val-Lys; Lys-Val; Ala-Lys; Lys-Ala; Phe-Cit; Cit-Phe; Leu-Cit; Cit-Leu; Ile-Cit; Cit-Ile; Phe-Arg; Arg-Phe; Cit-Trp; and Trp-Cit, or a pharmaceutically acceptable salt thereof.
  • linkers according to structural formula (IVa) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an antibody):
  • linkers according to structural formula (IVb), (IVc), or (IVd) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an antibody):
  • the linker comprises an enzymatically cleavable sugar moiety, for example, a linker comprising structural formula (Va), (Vb), (Vc), (Vd), or (Ve)::
  • q is 0 or 1
  • r is 0 or 1;
  • X 1 is CH 2 , O or NH
  • linkers according to structural formula (Va) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an anti-B7-H3 antibody):
  • linkers according to structural formula (Vb) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an anti-B7-H3 antibody):
  • linkers according to structural formula (Vc) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an anti-B7-H3 antibody):
  • linkers according to structural formula (Vd) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an anti-B7-H3 antibody):
  • linkers according to structural formula (Ve) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an anti-B7-H3 antibody):
  • the linkers comprising the ADC described herein need not be cleavable.
  • the drug release does not depend on the differential properties between the plasma and some cytoplasmic compartments.
  • the release of the drug is postulated to occur after internalization of the ADC via antigen-mediated endocytosis and delivery to lysosomal compartment, where the anti-B7-H3 antibody is degraded to the level of amino acids through intracellular proteolytic degradation. This process releases a drug derivative, which is formed by the drug, the linker, and the amino acid residue to which the linker was covalently attached.
  • Non-cleavable linkers may be alkylene chains, or maybe polymeric in natures, such as, for example, based upon polyalkylene glycol polymers, amide polymers, or may include segments of alkylene chains, polyalkylene glycols and/or amide polymers.
  • the linker comprises a polyethylene glycol segment having from 1 to 6 ethylene glycol units.
  • the linker is non-cleavable in vivo, for example a linker according to structural formula (VIa), (VIb), (VIc) or (VId) (as illustrated, the linkers include a group suitable for covalently linking the linker to an anti-B7-H3 antibody:
  • R a is selected from hydrogen, alkyl, sulfonate and methyl sulfonate;
  • R x is a moiety including a functional group capable of covalently linking the linker to an antibody;
  • linkers according to structural formula (VIa)-(VId) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an anti-B7-H3 antibody, and“ “ represents the point of attachment to a Bcl-xL inhibitor):
  • Attachment groups can be electrophilic in nature and include: maleimide groups, activated disulfides, active esters such as NHS esters and HOBt esters, haloformates, acid halides, alkyl and benzyl halides such as haloacetamides.
  • maleimide groups activated disulfides
  • active esters such as NHS esters and HOBt esters
  • haloformates acid halides
  • alkyl and benzyl halides such as haloacetamides.
  • the maleimide ring of a linker may react with an antibody Ab, forming a covalent attachment as either a succinimide (closed form) or succinamide (open form).
  • Polytherics has disclosed a method for bridging a pair of sulfhydryl groups derived from reduction of a native hinge disulfide bond. See, Badescu et al., 2014, Bioconjugate Chem.25:1124- 1136. The reaction is depicted in the schematic below.
  • An advantage of this methodology is the ability to synthesize homogenous DAR4 ADCs by full reduction of IgGs (to give 4 pairs of sulfhydryls) followed by reaction with 4 equivalents of the alkylating agent.
  • ADCs containing “bridged disulfides” are also claimed to have increased stability.
  • attachment moiety comprises the structural formulae (VIIa), (VIIb), or (VIIc):
  • R q is H or–O-(CH 2 CH 2 O) 11 -CH 3 ;
  • x is 0 or 1
  • y is 0 or 1
  • G 3 is–CH 2 CH 2 CH 2 SO 3 H or–CH 2 CH 2 O-(CH 2 CH 2 O) 11 -CH 3 ;
  • R w is–O-CH 2 CH 2 SO 3 H or–NH(CO)-CH 2 CH 2 O-(CH 2 CH 2 O) 12 -CH 3 ;
  • the linker comprises a segment according to structural formulae III III r III
  • R q is H or–O-(CH 2 CH 2 O) 11 -CH 3 ;
  • x is 0 or 1
  • y is 0 or 1
  • G 3 is–CH 2 CH 2 CH 2 SO 3 H or–CH 2 CH 2 O-(CH 2 CH 2 O) 11 -CH 3 ;
  • R w is–O-CH 2 CH 2 SO 3 H or–NH(CO)-CH 2 CH 2 O-(CH 2 CH 2 O) 12 -CH 3 ;
  • linkers according to structural formula (VIIa) and (VIIb) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an antibody):
  • linkers according to structural formula (VIIc) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an antibody):
  • L is selected from the group consisting of IVa.1-IVa.8, IVb.1- IVb.19, IVc.1-IVc.7, IVd.1-IVd.4, Va.1-Va.12, Vb.1-Vb.10, Vc.1-Vc.11, Vd.1-Vd.6, Ve.1-Ve.2, VIa.1, VIc.1-V1c.2, VId.1-VId.4, VIIa.1-VIIa.4, VIIb.1-VIIb.8, VIIc.1-VIIc.6 in either the closed or open form.
  • L is selected from the group consisting of IVb.2, IVc.5, IVc.6, IVc.7, IVd.4, Vb.9, Vc.11, VIIa.1, VIIa.3, VIIc.1, VIIc.4, and VIIc.5, wherein the maleimide of each linker has reacted with the antibody Ab, forming a covalent attachment as either a succinimide (closed form) or succinamide (open form).
  • linker L is selected from the group consisting of IVb.2, IVc.5, IVc.6, IVd.4, Vc.11, VIIa.1, VIIa.3, VIIc.1, VIIc.4, VIIc.5, wherein the maleimide of each linker has reacted with the antibody Ab, forming a covalent attachment as either a succinimide (closed form) or succinamide (open form).
  • linker L is selected from the group consisting of IVb.2, Vc.11, VIIa.3, IVc.6, and VIIc.1, wherein is the attachment point to drug D and @ is the attachment point to the LK, wherein when the linker is in the open form as shown below, @ can be either at the ⁇ - position or ⁇ -position of the carboxylic acid next to it:
  • the linker selected for a particular ADC may be influenced by a variety of factors, including but not limited to, the site of attachment to the antibody (e.g., lys, cys or other amino acid residues), structural constraints of the drug pharmacophore and the lipophilicity of the drug.
  • the specific linker selected for an ADC should seek to balance these different factors for the specific antibody/drug combination.
  • ADCs have been observed to effect killing of bystander antigen-negative cells present in the vicinity of the antigen-positive tumor cells.
  • the mechanism of bystander cell killing by ADCs has indicated that metabolic products formed during intracellular processing of the ADCs may play a role.
  • Neutral cytotoxic metabolites generated by metabolism of the ADCs in antigen-positive cells appear to play a role in bystander cell killing while charged metabolites may be prevented from diffusing across the membrane into the medium and therefore cannot affect bystander killing.
  • the linker is selected to attenuate the bystander killing effect caused by cellular metabolites of the ADC.
  • the linker is selected to increase the bystander killing effect.
  • the properties of the linker may also impact aggregation of the ADC under conditions of use and/or storage.
  • ADCs reported in the literature contain no more than 3-4 drug molecules per antibody molecule (see, e.g., Chari, 2008, Acc Chem Res 41:98-107).
  • DAR drug-to-antibody ratios
  • the linker incorporates chemical moieties that reduce aggregation of the ADCs during storage and/or use.
  • a linker may incorporate polar or hydrophilic groups such as charged groups or groups that become charged under physiological pH to reduce the aggregation of the ADCs.
  • a linker may incorporate charged groups such as salts or groups that deprotonate, e.g., carboxylates, or protonate, e.g., amines, at physiological pH.
  • the aggregation of the ADCs during storage or use is less than about 40% as determined by size-exclusion chromatography (SEC). In particular embodiments, the aggregation of the ADCs during storage or use is less than 35%, such as less than about 30%, such as less than about 25%, such as less than about 20%, such as less than about 15%, such as less than about 10%, such as less than about 5%, such as less than about 4%, or even less, as determined by size- exclusion chromatography (SEC).
  • SEC size-exclusion chromatography
  • Antibody-Drug Conjugate synthons are synthetic intermediates used to form ADCs.
  • the synthons are generally compounds according to structural formula (III):
  • the ADC synthons are compounds according to structural formulae (IIIa) and (IIIb) , or salts thereof, where the various substituents are as previously defined for structural formulae (IIa) and (IIb), respectively, and L and R x are as defined for structural formula (III):
  • an intermediate synthon according to structural formula (III), or a salt thereof is contacted with an antibody of interest under conditions in which functional group R x reacts with a“complementary” functional group on the antibody, F x , to form a covalent linkage.
  • groups R x and F x will depend upon the chemistry used to link the synthon to the antibody. Generally, the chemistry used should not alter the integrity of the antibody, for example its ability to bind its target. Preferably, the binding properties of the conjugated antibody will closely resemble those of the unconjugated antibody.
  • a variety of chemistries and techniques for conjugating molecules to biological molecules such as antibodies are known in the art and in particular to antibodies, are well-known. See, e.g., Amon et al.,“Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy,” in: Monoclonal Antibodies And Cancer Therapy, Reisfeld et al., Eds., Alan R.
  • R x comprises a functional group capable of linking the synthon to an amino group on an antibody.
  • R x comprises an NHS-ester or an
  • R x comprises a functional group capable of linking the synthon to a sulfhydryl group on an antibody.
  • R x comprises a haloacetyl or a maleimide.
  • L is selected from IVa or IVb and salts thereof; and R x comprises a functional group selected from the group consisting of NHS-ester, isothiocyanate, haloacetyl and maleimide.
  • the synthons are linked to the side chains of amino acid residues of the antibody, including, for example, the primary amino group of accessible lysine residues or the sulfhydryl group of accessible cysteine residues.
  • Free sulfhydryl groups may be obtained by reducing interchain disulfide bonds.
  • LK is a linkage formed with an amino group on the anti-B7-H3 antibody Ab (e.g., huAb13v1, huAb3v2.5, or huAb3v2.6).
  • LK is an amide or a thiourea.
  • LK is a linkage formed with a sulfhydryl group on the anti-B7-H3 antibody Ab.
  • LK is a thioether.
  • LK is selected from the group consisting of amide, thiourea and thioether; and m is an integer ranging from 1 to 8.
  • R x and chemistries useful for linking synthons to accessible lysine residues are known, and include by way of example and not limitation NHS-esters and isothiocyanates.
  • a number of functional groups R x and chemistries useful for linking synthons to accessible free sulfhydryl groups of cysteine residues are known, and include by way of example and not limitation haloacetyls and maleimides.
  • conjugation chemistries are not limited to available side chain groups.
  • Side chains such as amines may be converted to other useful groups, such as hydroxyls, by linking an appropriate small molecule to the amine.
  • This strategy can be used to increase the number of available linking sites on the antibody by conjugating multifunctional small molecules to side chains of accessible amino acid residues of the antibody.
  • Functional groups R x suitable for covalently linking the synthons to these“converted” functional groups are then included in the synthons.
  • the antibody may also be engineered to include amino acid residues for conjugation.
  • An approach for engineering antibodies to include non-genetically encoded amino acid residues useful for conjugating drugs in the context of ADCs is described in Axup et al., 2003, Proc Natl Acad Sci 109:16101-16106 and Tian et al., 2014, Proc Natl Acad Sci 111:1776-1771, as are chemistries and functional group useful for linking synthons to the non-encoded amino acids.
  • Exemplary synthons useful for making ADCs described herein include, but are not limited to, the following synthons listed below in Table B.
  • the synthon is selected from the group consisting of synthon examples 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 2.10, 2.11, 2.12, 2.13, 2.14, 2.15, 2.16, 2.17, 2.18, 2.19, 2.20, 2.21, 2.22, 2.23, 2.24, 2.25, 2.26, 2.27, 2.28, 2.29, 2.30, 2.31, 2.34, 2.35, 2.36, 2.37, 2.38, 2.39, 2.40, 2.41, 2.42, 2.43, 2.44, 2.45, 2.46, 2.47, 2.48, 2.49, 2.50, 2.51, 2.52, 2.53, 2.54, 2.55, 2.56, 2.57, 2.58, 2.59, 2.60, 2.61, 2.62, 2.63, 2.64, 2.65, 2.66, 2.67, 2.68, 2.69, 2.70, 2.71, 2.72, and pharmaceutically acceptable salts thereof.
  • the corresponding compound names of these synthons are provided

Abstract

The invention relates to B7 homology 3 protein (B7-H3) antibodies and antibody drug conjugates (ADCs), including compositions and methods of using said antibodies and ADCs.

Description

ANTI-B7-H3 ANTIBODIES AND ANTIBODY DRUG CONJUGATES RELATED APPLICATIONS
This application claims priority to U.S. Provisional Application No.62/347,394, filed on June 8, 2016, and to U.S. Provisional Application No.62/366,478, filed on July 25, 2016. The entire contents of the foregoing applications are expressly incorporated herein by reference. SEQUENCE LISTING
The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on June 7, 2017, is named 117813-10620_ST25.txt and is 159,744 bytes in size. BACKGROUND OF THE INVENTION
The B7 homology 3 protein (B7-H3) (also known as CD276 and B7RP-2, and referred to herein as“B7-H3”) is a type I transmembrane glycoprotein of the immunoglobulin superfamily. Human B7-H3 contains a putative signal peptide, V-like and C-like Ig domains, a transmembrane region and a cytoplasmic domain. Exon duplication in humans results in the expression of two B7-H3 isoforms having either a single IgV-IgC-like domain (2IgB7-H3 isoform) or a IgV-IgC-IgV-IgC-like domain (4IgB7-H3 isoform) containing several conserved cysteine residues. The predominant B7-H3 isoform in human tissues and cell lines is the 4IgB7-H3 isoform (Steinberger et al., J. Immunol. 172(4): 2352-9 (2004)).
B7-H3 has been reported as having both co-stimulatory and co-inhibitory signaling functions (see, e.g., Chapoval et al., Nat. Immunol.2: 269-74 (2001); Suh et al., Nat. Immunol.4: 899-906 (2003); Prasad et al., J. Immunol.173: 2500-6 (2004); and Wang et al., Eur. J. Immunol.35: 428-38 (2005)). For example, in vitro studies have shown B7-H3’s co-stimulatory function since B7-H3 was able to increase proliferation of cytotoxic T-lymphocytes (CTLs) and upregulate interferon gamma (IFN-γ) production in the presence of anti-CD3 antibody to mimic the T cell receptor signal
(Chapoval et al., 2001). Moreover, in vivo studies using cardiac allografts in B7-H3 -/- mice showed decreased production of key cytokine, chemokine and chemokine receptor mRNA transcripts (e.g., IL-2, IFN-γ, monocyte chemoattractant protein (MCP-1) and IFN-inducible protein (IP)-10) as compared to wild-type control (Wang et al., 2005). In contrast, B7-H3 co-inhibitory function has been observed, for example, in mice where B7-H3 protein inhibited T-cell activation and effector cytokine production (Suh et al., 2003). Although no ligands have been identified for human B7-H3, murine B7-H3 has been found to bind to the triggering receptor expressed on myeloid cells (TREM-) like transcript 2 (TLT-2), a modulator of adaptive an innate immunity cellular responses. Binding of murine B7-H3 to TLT-2 on CD8+ T-cells enhances T-cell effector functions such as proliferation, cytotoxicity and cytokine production (Hashiguchi et al., Proc. Nat'l. Acad. Sci. U.S.A. 105(30): 10495-500 (2008)).
B7-H3 is not constitutively expressed in many immune cells {e.g., natural killer (NK) cells, T- cells, and antigen-presenting cells (APCs)); however, its expression can be induced. Further, the expression of B7-H3 is not restricted to immune cells. B7-H3 transcripts are expressed in a variety of human tissues including colon, heart, liver, placenta, prostate, small intestine, testis, and uterus, as well as osteoblasts, fibroblasts, epithelial cells, and other cells of non-lymphoid lineage, potentially indicating immunological and non-immunological functions (Nygren et al. Front Biosci. 3:989-93 (2011)). However, protein expression in normal tissue is typically maintained at a low level and thus, may be subject to post-transcriptional regulation.
B7-H3 is also expressed in a variety of human cancers, including prostate cancer, clear cell renal cell carcinoma, glioma, melanoma, lung cancer, non-small cell lung cancer (NSCLC), small cell lung cancer, pancreatic cancer, gastric cancer, acute myeloid leukemia (AML), non-Hodgkin's lymphoma (NHL), ovarian cancer, colorectal cancer, colon cancer, renal cancer, hepatocellular carcinoma, kidney cancer, head and neck cancer, hypopharyngeal squamous cell carcinoma, glioblastoma, neuroblastoma, breast cancer, endometrial cancer, and urothelial cell carcinoma.
Although the role of B7-H3 in cancer cells is unclear, its expression may orchestrate signaling events that may protect cancer cells from innate and adaptive immune responses. For example, B7-H3 is overexpressed in high-grade prostatic intraepithelial neoplasia and adenocarcinomas of the prostate, and high expression levels of B7-H3 in these cancerous cells is associated with an increased risk of cancer progression after surgery (Roth et al. Cancer Res. 67(16): 7893-900 (2007)). Further, tumor B7-H3 expression in NSCLC inversely correlated with the number of tumor-infiltrating lymphocytes and significantly correlated with lymph node metastasis (Sun et al. Lung Cancer 53(2): 143-51 (2006)). The level of circulating soluble B7-H3 in NSCLC patients has also been associated with higher tumor stage, tumor size, lymph node metastasis, and distant metastasis (Yamato et al., Br. J. Cancer 101(10): 1709-16 (2009)).
B7-H3 may also play an important role in T-cell-mediated antitumor responses in a context dependent manner. For example, gastric cancer tumor cell expression of B7-H3 positively correlated with survival time, infiltration depth, and tissue type (Wu et al., World J. Gastroenterol. 12(3): 457-9 (2006)). Further, high expression of B7-H3 in pancreatic tumor cells was associated with patient survival after surgical resection and significantly correlated with the number of tumor-infiltrating CD8+ T-cells (Loos et al, BMC Cancer 9:463 (2009).
Antibody drug conjugates (ADC) represent a relatively new class of therapeutics comprising an antibody conjugated to a cytotoxic drug via a chemical linker. The therapeutic concept of ADCs is to combine binding capabilities of an antibody with a drug, where the antibody is used to deliver the drug to a tumor cell by means of binding to a target surface antigen, including target surface antigens that are overexpressed in the tumor cells. There remains a need in the art for anti-B7-H3 antibodies and anti-B7-H3 ADCs that can be used for therapeutic purposes in the treatment of cancer. SUMMARY OF THE INVENTION
In certain aspects, the present invention provides for antibodies and antibody drug conjugates (ADCs) that specifically bind to human B7-H3. In certain aspects, the present invention provides novel ADCs that can selectively deliver Bcl-xL inhibitors to target cancer cells, e.g., B7-H3 expressing cells.
In one aspect, the present invention provide an anti-B7H3 antibody, or antigen binding portion thereof, that binds to human B7-H3 (hB7-H3), wherein the anti-B7H3 antibody, or antigen binding portion thereof, comprises a heavy chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 12 and a light chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 15. In one embodiment, the anti-B7H3 antibody, or antigen binding portion thereof, comprises a heavy chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 140 and a light chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 7. In one embodiment, the anti-B7H3 antibody, or antigen binding portion thereof, comprises a heavy chain variable region comprising a CDR1 having the amino acid sequence of SEQ ID NO: 10 and a light chain variable region comprising a CDR1 having the amino acid sequence of either SEQ ID NO: 136 or 138.
In one aspect, the present invention provides an anti-B7H3 antibody antibody, or antigen binding portion thereof, that binds to human B7-H3, wherein the anti-B7H3 antibody, or antigen binding portion thereof, comprises a heavy chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 35 and a light chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 39. In one embodiment, the anti-B7H3 antibody, or antigen binding portion thereof, comprises a heavy chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 34, and a light chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 38. In one embodiment, the anti-B7H3 antibody, or antigen binding portion thereof, comprises a heavy chain variable region comprising a CDR1 having the amino acid sequence of SEQ ID NO: 33 and a light chain variable region comprising a CDR1 having the amino acid sequence of either SEQ ID NO: 37.
In one embodiment, the anti-B7H3 antibody, or antigen binding portion thereof, is an IgG isotype.
In one embodiment, the anti-B7H3 antibody, or antigen binding portion thereof, is an IgG1 or an IgG4 isotype.
In one embodiment, the anti-B7H3 antibody, or antigen binding portion thereof, has a KD of 1.5 x 10-8 or less as determined by surface plasmon resonance. In one aspect, the present invention provides an anti-B7H3 antibody, or antigen-binding portion thereof, that binds to hB7-H3, said anti-B7H3 antibody, or antigen-binding portion thereof, comprising a heavy chain variable region comprising either a CDR set of SEQ ID NOs: 10, 11, and 12, and a light chain variable region comprising a CDR set of SEQ ID NOs: 14, 7, and 15; or a heavy chain variable region comprising a CDR set of SEQ ID NOs: 33, 35, and 35, and a light chain variable region comprising a CDR set of SEQ ID NOs: 37, 38, and 39. In one embodiment, the anti-B7H3 antibody, or antigen binding portion thereof, is humanized. In one embodiment, the anti-B7H3 antibody, or antigen binding portion thereof, further comprises a human acceptor framework. In one embodiment, the human acceptor framework comprises an amino acid sequence selected from the group consisting of SEQ ID Nos: 155, 156, 164, 165, 166, and 167. In one embodiment, the human acceptor framework comprises at least one framework region amino acid substitution. In one embodiment, the amino acid sequence of the framework is at least 65% identical to the sequence of the human acceptor framework and comprises at least 70 amino acid residues identical to the human acceptor framework. In one embodiment, the amino acid sequence of the framework is at least 85% identical, 90% identical, 95% identical, 96% identical, 97% identical, 98% identical, or 99% identical to the sequence of the human acceptor framework and comprises at least 70, at least 75, at least 80, or at least 85 amino acid residues identical to the human acceptor framework.
In one embodiment, the human acceptor framework comprises at least one framework region amino acid substitution at a key residue, said key residue selected from the group consisting of: a residue adjacent to a CDR; a glycosylation site residue; a rare residue; a residue capable of interacting with human B7-H3; a residue capable of interacting with a CDR; a canonical residue; a contact residue between heavy chain variable region and light chain variable region; a residue within a Vernier zone; and a residue in a region that overlaps between a Chothia-defined variable heavy chain CDR1 and a Kabat-defined first heavy chain framework. In one embodiment, the key residue is selected from the group consisting of 48H, 67H, 69H, 71H, 73H, 94H, and 2L (H refers to the heavy chain; L refers to the light chain; amino acid residues in reference to the Kabat numbering system). In one embodiment, the key residue substitution is in the variable heavy chain region and is selected from the group consisting of M48I, V67A, I69L, A71V, K73R, and R94G. In one embodiment, the key residue substitution is in the variable light chain region and is I2V.
In one aspect, the present invention provides an anti-B7H3 antibody, or antigen-binding portion thereof, that binds to hB7-H3 comprising a heavy chain variable region comprising a CDR set of SEQ ID NOs: 25, 26, and 27, and a light chain variable region comprising a CDR set of SEQ ID NOs: 29, 30, and 31. In one embodiment, the anti-B7H3 antibody, or antigen binding portion thereof, is humanized. In one embodiment, the antibody, or antigen binding portion thereof, further comprises a human acceptor framework. In one embodiment, the human acceptor framework comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 155 to 158. In one aspect, the present invention provides an anti-B7H3 antibody, or antigen-binding portion thereof, that binds to hB7-H3 comprising a heavy chain variable region comprising a CDR set of SEQ ID NOs: 33, 35, and 35, and a light chain variable region comprising a CDR set of SEQ ID NOs: 37, 38, and 39. In one embodiment, the anti-B7H3 antibody, or antigen binding portion thereof, is humanized. In one embodiment, the anti-B7H3 antibody, or antigen binding portion thereof, further comprises a human acceptor framework. In one embodiment, human acceptor framework comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 156, 158, 166 and 167.
In one embodiment, the human acceptor framework comprises at least one framework region amino acid substitution. In one embodiment, the amino acid sequence of the framework is at least 65% identical to the sequence of the human acceptor framework and comprises at least 70 amino acid residues identical to the human acceptor framework. In one embodiment, the amino acid sequence of the framework is at least 85% identical, 90% identical, 95% identical, 96% identical, 97% identical, 98% identical, or 99% identical to the sequence of the human acceptor framework and comprises at least 70, at least 75, at least 80, or at least 85 amino acid residues identical to the human acceptor framework.
In one embodiment, the human acceptor framework comprises at least one framework region amino acid substitution at a key residue, said key residue selected from the group consisting of: a residue adjacent to a CDR; a glycosylation site residue; a rare residue; a residue capable of interacting with human B7-H3; a residue capable of interacting with a CDR; a canonical residue; a contact residue between heavy chain variable region and light chain variable region; a residue within a Vernier zone; and a residue in a region that overlaps between a Chothia-defined variable heavy chain CDR1 and a Kabat-defined first heavy chain framework. In one embodiment, the key residue is selected from the group consisting of 69H, 46L, 47L, 64L, and 71L (H refers to the heavy chain; L refers to the light chain; amino acid residues in reference to the Kabat numbering system). In one embodiment, the key residue substitution is in the variable heavy chain region and is L69I. In one embodiment, the key residue substitution is in the variable light chain region and is selected from the group consisting of L46P, L47W, G64V, and F71H.
In one embodiment, the present invention provides an anti-hB7-H3 antibody, or antigen- binding portion thereof, comprising a heavy chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 10, a heavy chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 140, a heavy chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 12, a light chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 136 or 138, a light chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 7, and a light chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 15.
In another embodiment, the present invention provides an anti-hB7-H3 antibody, or antigen- binding portion thereof comprising a heavy chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 33, a heavy chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 34, a heavy chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 35, a light chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 37, a light chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 38, and a light chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 39.
In one embodiment, the present invention provides an anti-hB7-H3 antibody, or antigen- binding portion thereof, comprising a heavy chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 139 and a light chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 135.
In one embodiment, the present invention provides an anti-hB7-H3 antibody, or antigen- binding portion thereof, comprising a heavy chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 139, and/or a light chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 135.
In one embodiment, the present invention provides an anti-hB7-H3 antibody, or antigen- binding portion thereof, comprising a heavy chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 139 and a light chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 137.
In one embodiment, the present invention provides an anti-hB7-H3 antibody, or antigen- binding portion thereof, comprising a heavy chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 139, and/or a light chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 137.
In one embodiment, the present invention provides an anti-hB7-H3 antibody, or antigen- binding portion thereof, comprising a heavy chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 147 and a light chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 144.
In one embodiment, the present invention provides an anti-hB7-H3 antibody, or antigen- binding portion thereof, comprising a heavy chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 147, and/or a light chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 144.
In one aspect, the present invention provide an anti-B7H3 antibody that binds to human B7- H3 (hB7-H3), wherein the anti-B7H3 antibody comprises a heavy chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 12 and a light chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 15. In one embodiment, the anti-B7H3 antibody comprises a heavy chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 140 and a light chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 7. In one embodiment, the anti-B7H3 antibody comprises a heavy chain variable region comprising a CDR1 having the amino acid sequence of SEQ ID NO: 10 and a light chain variable region comprising a CDR1 having the amino acid sequence of either SEQ ID NO: 136 or 138.
In one aspect, the present invention provides an anti-B7H3 antibody antibody that binds to human B7-H3, wherein the anti-B7H3 antibody comprises a heavy chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 35 and a light chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 39. In one embodiment, the anti-B7H3 antibody comprises a heavy chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 34, and a light chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 38. In one embodiment, the anti-B7H3 antibody comprises a heavy chain variable region comprising a CDR1 having the amino acid sequence of SEQ ID NO: 33 and a light chain variable region comprising a CDR1 having the amino acid sequence of either SEQ ID NO: 37.
In one embodiment, the anti-B7H3 antibody is an IgG isotype.
In one embodiment, the anti-B7H3 antibody is an IgG1 or an IgG4 isotype.
In one embodiment, the anti-B7H3 antibody has a KD of 1.5 x 10-8 or less as determined by surface plasmon resonance.
In one aspect, the present invention provides an anti-B7H3 antibody that binds to hB7-H3, said anti-B7H3 antibody comprising a heavy chain variable region comprising either a CDR set of SEQ ID NOs: 10, 11, and 12, and a light chain variable region comprising a CDR set of SEQ ID NOs: 14, 7, and 15; or a heavy chain variable region comprising a CDR set of SEQ ID NOs: 33, 35, and 35, and a light chain variable region comprising a CDR set of SEQ ID NOs: 37, 38, and 39. In one embodiment, the anti-B7H3 antibody is humanized. In one embodiment, the anti-B7H3 antibody further comprises a human acceptor framework. In one embodiment, the human acceptor framework comprises an amino acid sequence selected from the group consisting of SEQ ID Nos: 155, 156, 164, 165, 166, and 167. In one embodiment, the human acceptor framework comprises at least one framework region amino acid substitution. In one embodiment, the amino acid sequence of the framework is at least 65% identical to the sequence of the human acceptor framework and comprises at least 70 amino acid residues identical to the human acceptor framework. In one embodiment, the amino acid sequence of the framework is at least 85% identical, 90% identical, 95% identical, 96% identical, 97% identical, 98% identical, or 99% identical to the sequence of the human acceptor framework and comprises at least 70, at least 75, at least 80, or at least 85 amino acid residues identical to the human acceptor framework.
In one aspect, the present invention provides an anti-B7H3 antibody that binds to hB7-H3 comprising a heavy chain variable region comprising a CDR set of SEQ ID NOs: 25, 26, and 27, and a light chain variable region comprising a CDR set of SEQ ID NOs: 29, 30, and 31. In one embodiment, the anti-B7H3 antibody is humanized. In one embodiment, the antibody further comprises a human acceptor framework. In one embodiment, the human acceptor framework comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 155 to 158.
In one aspect, the present invention provides an anti-B7H3 antibody that binds to hB7-H3 comprising a heavy chain variable region comprising a CDR set of SEQ ID NOs: 33, 35, and 35, and a light chain variable region comprising a CDR set of SEQ ID NOs: 37, 38, and 39. In one embodiment, the anti-B7H3 antibod is humanized. In one embodiment, the anti-B7H3 antibody further comprises a human acceptor framework. In one embodiment, human acceptor framework comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 156, 158, 166 and 167.
In one embodiment, the human acceptor framework comprises at least one framework region amino acid substitution at a key residue, said key residue selected from the group consisting of: a residue adjacent to a CDR; a glycosylation site residue; a rare residue; a residue capable of interacting with human B7-H3; a residue capable of interacting with a CDR; a canonical residue; a contact residue between heavy chain variable region and light chain variable region; a residue within a Vernier zone; and a residue in a region that overlaps between a Chothia-defined variable heavy chain CDR1 and a Kabat-defined first heavy chain framework. In one embodiment, the key residue is selected from the group consisting of 69H, 46L, 47L, 64L, and 71L (H refers to the heavy chain; L refers to the light chain; amino acid residues in reference to the Kabat numbering system). In one embodiment, the key residue substitution is in the variable heavy chain region and is L69I. In one embodiment, the key residue substitution is in the variable light chain region and is selected from the group consisting of L46P, L47W, G64V, and F71H.
In one embodiment, the present invention provides an anti-hB7-H3 antibody comprising a heavy chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 10, a heavy chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 140, a heavy chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 12, a light chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 136 or 138, a light chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 7, and a light chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 15.
In another embodiment, the present invention provides an anti-hB7-H3 antibody comprising a heavy chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 33, a heavy chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 34, a heavy chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 35, a light chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 37, a light chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 38, and a light chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 39. In one embodiment, the present invention provides an anti-hB7-H3 antibody comprising a heavy chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 139 and a light chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 135.
In one embodiment, the present invention provides an anti-hB7-H3 antibody comprising a heavy chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 139, and/or a light chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 135.
In one embodiment, the present invention provides an anti-hB7-H3 antibody comprising a heavy chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 139 and a light chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 137.
In one embodiment, the present invention provides an anti-hB7-H3 antibody comprising a heavy chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 139, and/or a light chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 137.
In one embodiment, the present invention provides an anti-hB7-H3 antibody comprising a heavy chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 147 and a light chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 144.
In one embodiment, the present invention provides an anti-hB7-H3 antibody comprising a heavy chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 147, and/or a light chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 144.
In one embodiment, the antibody, or antigen-binding portion thereof, provided herein binds to cynomolgus (cyno) B7-H3.
In one embodiment, the antibody, or antigen binding portion thereof, has a dissociation constant (KD) to hB7-H3 selected from the group consisting of: at most about 10-7 M; at most about 10-8 M; at most about 10-9 M; at most about 10-10 M; at most about 10-11 M; at most about 10-12 M; and at most 10-13 M.
In one embodiment, the antibody, or antigen binding portion thereof, comprises a heavy chain immunoglobulin constant domain of a human IgM constant domain, a human IgG1 constant domain, a human IgG2 constant domain, a human IgG3 constant domain, a human IgG4 constant domain, a human IgA constant domain, or a human IgE constant domain. In one embodiment, the antibody is an IgG1 monoclonal antibody comprising a kappa light chain. In one embodiment, the human IgG1 constant domain comprises an amino acid sequence of SEQ ID NO: 159 or SEQ ID NO: 160.
In one aspect, the present invention provides an anti-hB7-H3 antibody comprising a sequence set selected from the group consisting of: a) a heavy chain comprising the amino acid sequence of SEQ ID NO: 168 and a light chain comprising the amino acid sequence of SEQ ID NO: 169; b) a heavy chain comprising the amino acid sequence of SEQ ID NO: 170 and a light chain comprising the amino acid sequence of SEQ ID NO: 171; and c) a heavy chain comprising the amino acid sequence of SEQ ID NO: 172 and a light chain comprising the amino acid sequence of SEQ ID NO: 173.
In one embodiment, the anti-hB7-H3 antibody, or antigen-binding portion thereof, comprises a heavy chain CDR set corresponding to antibody huAb13v1, and a light chain CDR set
corresponding to antibody huAb13v1. In one embodiment, the anti-hB7-H3 antibody, or antigen- binding portion thereof, comprises a heavy chain variable region corresponding to antibody huAb13v1, and a light chain variable region corresponding to antibody huAb13v1.
In one embodiment, the anti-hB7-H3 antibody, or antigen-binding portion thereof, comprises a heavy chain CDR set corresponding to antibody huAb3v2.5, and a light chain CDR set
corresponding to antibody huAb3v2.5. In one embodiment, the anti-hB7-H3 antibody, or antigen- binding portion thereof, comprises a heavy chain variable region corresponding to antibody huAb3v2.5, and a light chain variable region corresponding to antibody huAb3v2.5.
In one embodiment, the anti-hB7-H3 antibody, or antigen-binding portion thereof, competes with the antibody, or antigen binding portion thereof, of any one of the anti-hB7-H3 antibodies, or antigen-binding portions thereof, disclosed herein.
In one embodiment, the anti-hB7-H3 antibody is an IgG, e.g., and IgG1, having four polypeptide chains which are two heavy chains and two light chains.
In one aspect, the present invention provides a pharmaceutical composition comprising an anti-hB7-H3 antibody, or antigen binding portion thereof, as disclosed herein, and a pharmaceutically acceptable carrier.
In another aspect, the present invention provides an anti-hB7-H3 Antibody Drug Conjugate (ADC) comprising an anti-hB7-H3 antibody disclosed herein conjugated to a drug via a linker. In one embodiment, the drug is an auristatin or a pyrrolobenzodiazepine (PBD). In one embodiment, the drug is a Bcl-xL inhibitor.
In one aspect, the present invention provides an anti-hB7-H3 antibody drug conjugate (ADC) comprising a drug linked to an anti-human B7-H3 (hB7-H3) antibody by way of a linker, wherein the drug is a Bcl-xL inhibitor according to structural formula (IIa) or (IIb):
Figure imgf000011_0001
Figure imgf000012_0001
,
Figure imgf000012_0002
ly selected from halo h drox nitro lower alk l lower heteroalk l C1-4alkox amino c ano and
Figure imgf000012_0003
independently selected from halo, hydroxy, nitro, lower alkyl, lower heteroalkyl, C1-4alkoxy, amino, cyano and halomethyl, wherein the #-N(R4)-R13-Z2b- substituent of formula (IIb) is attached to Ar2 at any Ar2 atom capable of being substituted; Z1 is selected from N, CH, C-halo and C-CN; Z2a, Z2b, and Z2c are each, independent from one another, selected from a bond, NR6, CR6aR6b, O, S, S(O), SO2, NR6C(O), NR6aC(O)NR6b, and NR6C(O)O; R1 is selected from hydrogen, methyl, halo, halomethyl, ethyl and cyano; R2 is selected from hydrogen, methyl, halo, halomethyl and cyano; R3 is selected from hydrogen, lower alkyl and lower heteroalkyl; R4 is selected from hydrogen, lower alkyl, monocyclic cycloalkyl, monocyclic heterocyclyl, and lower heteroalkyl or is taken together with an atom of R13 to form a cycloalkyl or heterocyclyl ring having between 3 and 7 ring atoms, wherein the lower alkyl, monocyclic cycloalkyl, monocyclic heterocyclyl, and lower heteroalkyl are optionally substituted with one or more halo, cyano, hydroxy, C1-4alkoxy, monocyclic cycloalkyl, monocyclic heterocyclyl, C(O)NR6aR6b, S(O)2NR6aR6b, NHC(O)CHR6aR6b, NHS(O)CHR6aR6b,
NHS(O)2CHR6aR6b, S(O)2CHR6aR6b or S(O)2NH2 groups; R6, R6a and R6b are each, independent from one another, selected from hydrogen, lower alkyl, lower heteroalkyl, optionally substituted monocyclic cycloalklyl and monocyclic heterocyclyl, or are taken together with an atom from R13 to form a cycloalkyl or heterocyclyl ring having between 3 and 7 ring atoms; R10 is selected from cyano, OR14, SR14, SOR14, SO2R14, SO2NR14aR14b, NR14aR14b, NHC(O)R14 and NHSO2R14; R11a and R11b are each, independently of one another, selected from hydrogen, halo, methyl, ethyl, halomethyl, hydroxyl, methoxy, CN, and SCH3; R12 is selected from hydrogen, halo, cyano, lower alkyl, lower heteroalkyl, cycloalkyl, and heterocyclyl, wherein the alkyl, heteroalkyl, cycloalkyl, and heterocyclyl are optionally substituted with one or more halo, cyano, C1-4alkoxy, monocyclic cycloalkyl, monocyclic heterocyclyl, NHC(O)CHR6aR6b, NHS(O)CHR6aR6b, NHS(O)2CHR6aR6b or
S(O)2CHR6aR6b groups; R13 is selected from a bond, optionally substituted lower alkylene, optionally substituted lower heteroalkylene, optionally substituted cycloalkyl or optionally substituted heterocyclyl; R14 is selected from hydrogen, optionally substituted lower alkyl and optionally substituted lower heteroalkyl; R14a and R14b are each, independently of one another, selected from hydrogen, optionally substituted lower alkyl, and optionally substituted lower heteroalkyl, or are taken together with the nitrogen atom to which they are bonded to form an optionally substituted monocyclic cycloalkyl or monocyclic heterocyclyl ring; R15 is selected from hydrogen, halo, C1-6 alkanyl, C2-4 alkenyl, C2-4 alkynyl, and C1-4 haloalkyl and C1-4 hydroxyalkyl, with the proviso that when R15 is present, R4 is not C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C1-4 haloalkyl or C1-4 hydroxyalkyl, wherein the R4 C1-6 alkanyl, C2-4 alkenyl, C2-4 alkynyl, C1-4 haloalkyl and C1-4 hydroxyalkyl are optionally substituted with one or more substituents independently selected from OCH3,
OCH2CH2OCH3, and OCH2CH2NHCH3; and # represents a point of attachment to a linker; and wherein the anti-hB7-H3 antibody either: comprises a heavy chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 10, a heavy chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 140, a heavy chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 12, a light chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 136 or 138, a light chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 7, and a light chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 15; or comprises a heavy chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 33, a heavy chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 34 a heavy chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 35, a light chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 37, a light chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 38, and a light chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 39.
In one embodiment, the ADC is a compound according to structural formula (I):
Figure imgf000014_0001
wherein D is the Bcl-xL inhibitor drug of formula (IIa) or (IIb); L is the linker; Ab is the anti-hB7-H3 antibody; LK represents a covalent linkage linking the linker (L) to the anti-hB7-H3 antibody (Ab); and m is an integer ranging from 1 to 20.
In one embodiment, the Ar1 is unsubstituted. In one embodiment, the Ar1 is
In one embodiment, the Ar2 is unsubstituted. In one embodiment, the Ar2 is
Figure imgf000014_0002
, which yl, C1-4 alkoxy, and
cyano;
Figure imgf000014_0003
.
In one embodiment, Z1 is N.
In one embodiment, Z2a is O.
In one embodiment, R1 is methyl or chloro.
In one embodiment, R2 is hydrogen or methyl. In one embodiment, R2 is hydrogen.
In one embodiment, R4 is hydrogen or lower alkyl, wherein the lower alkyl is optionally substituted with C1-4 alkoxy or C(O)NR6aR6b.
Figure imgf000014_0004
optionally substituted at the 5-position with a group selected from hydroxyl, C1-4 alkoxy, and cyano.
In one embodiment, the drug is a Bcl-xL inhibitor according to structural formula (IIa). In one embodiment, the drug is a Bcl-xL inhibitor according to structural formula (IIa). In one embodiment, Z2a is CH2 or O. In one embodiment, R13 is selected from lower alkylene or lower heteroalkylene.
In one embodiment, the group
Figure imgf000015_0001
or
.
In one embodiment, the group .
In one embodiment, the group
Figure imgf000015_0002
,
Figure imgf000015_0003
.
In one embodiment, the group
Figure imgf000015_0004
. In one embodiment, Z2a is oxygen, R13 is CH2CH2, R4 is hydrogen or lower alkyl optionally substituted with C1-4 alkoxy or C(O)NR6aR6b.
In one embodiment, the ADC is a compound according to structural formula (IIb).
In one embodiment, Z2b is a bond, O, or NR6, or and R13 is ethylene or optionally substituted heterocyclyl.
In one embodiment, Z2c is O and R12 is lower alkyl optionally substituted with one or more halo or C1-4 alkoxy.
In one embodiment, the Bcl-xL inhibitor is selected from the group consisting of the following compounds modified in that the hydrogen corresponding to the # position of structural formula (IIa) or (IIb) is not present forming a monoradical: 6-[1-(1,3-benzothiazol-2-ylcarbamoyl)- 1,2,3,4-tetrahydroquinolin-7-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid; 6-[4-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-2H-1,4-benzoxazin-6-yl]-3-[1- ({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol- 4-yl]pyridine-2-carboxylic acid; 6-[4-(1,3-benzothiazol-2-ylcarbamoyl)-1-methyl-1,2,3,4- tetrahydroquinoxalin-6-yl]-3-[1-({35-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[33113,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid; 3-(1-{[3-(2-aminoethoxy)-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[1-(1,3-benzothiazol-2- ylcarbamoyl)-5,6-dihydroimidazo[1,5-a]pyrazin-7(8H)-yl]pyridine-2-carboxylic acid; 3-(1-{[3-(2- aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-5-hydroxy-3,4-dihydroisoquinolin-2(1H)-yl]pyridine-2-carboxylic acid; 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid; 3-[1-({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)- 5-methyl-1H-pyrazol-4-yl]-6-[8-([1,3]thiazolo[5,4-b]pyridin-2-ylcarbamoyl)naphthalen-2-yl]pyridine- 2-carboxylic acid; 3-[1-({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]-6-[8-([1,3]thiazolo[4,5-b]pyridin-2-ylcarbamoyl)naphthalen-2- yl]pyridine-2-carboxylic acid; 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-3-[1-({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec- 1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid; 6-[5-(1,3-benzothiazol-2- ylcarbamoyl)quinolin-3-yl]-3-[1-({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid; 6-[4-(1,3-benzothiazol-2- ylcarbamoyl)quinolin-6-yl]-3-[1-({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid; 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-3-{1-[(3-{2-[(2- methoxyethyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol- 4-yl}pyridine-2-carboxylic acid; 3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-cyano-3,4- dihydroisoquinolin-2(1H)-yl]pyridine-2-carboxylic acid; 6-[1-(1,3-benzothiazol-2-ylcarbamoyl)- 1,2,3,4-tetrahydroquinolin-7-yl]-3-{1-[(3-{2-[(2-methoxyethyl)amino]ethoxy}-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol-4-yl}pyridine-2-carboxylic acid; 6- [8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-3-{1-[(3-{2-[(2-methoxyethyl)amino]ethoxy}- 5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol-4-yl}pyridine-2-carboxylic acid; 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-[1-({3,5-dimethyl-7- [2-(oxetan-3-ylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine- 2-carboxylic acid; 6-[6-(3-aminopyrrolidin-1-yl)-8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]-3-(1-{[3-(2-methoxyethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid; 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-{1-[(3,5-dimethyl-7-{2-[(2- sulfamoylethyl)amino]ethoxy}tricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol-4- yl}pyridine-2-carboxylic acid; 3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[3-(1,3-benzothiazol-2-ylcarbamoyl)-6,7-dihydrothieno[3,2- c]pyridin-5(4H)-yl]pyridine-2-carboxylic acid; 3-(1-{[3-(2-aminoethoxy)-57- dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[1-(1,3-benzothiazol-2- ylcarbamoyl)-3-(trifluoromethyl)-5,6-dihydroimidazo[1,5-a]pyrazin-7(8H)-yl]pyridine-2-carboxylic acid; 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-6-{methyl[2-(methylamino)ethyl]amino}-3,4- dihydroisoquinolin-2(1H)-yl]-3-(1-{[3-(2-methoxyethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid; 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-6-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid; 3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl- 1H-pyrazol-4-yl)-6-[4-(1,3-benzothiazol-2-ylcarbamoyl)quinolin-6-yl]pyridine-2-carboxylic acid; 6- [5-amino-8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-[1-({3,5-dimethyl- 7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid; 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-6-[3-(methylamino)prop-1-yn-1-yl]-3,4- dihydroisoquinolin-2(1H)-yl]-3-(1-{[3-(2-methoxyethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid; 6-[4-(1,3-benzothiazol-2- ylcarbamoyl)isoquinolin-6-yl]-3-[1-({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec- 1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid; 6-[7-(1,3-benzothiazol-2- ylcarbamoyl)-1H-indol-2-yl]-3-[1-({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid; 3-(1-{[3-(2-aminoethoxy)-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[7-(1,3-benzothiazol-2- ylcarbamoyl)-1H-indol-2-yl]pyridine-2-carboxylic acid; 6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-3- methyl-1H-indol-2-yl]-3-[1-({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid; 6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-(1-{[3,5-dimethyl-7-(2-{[1- (methylsulfonyl)piperidin-4-yl]amino}ethoxy)tricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)pyridine-2-carboxylic acid; 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]-3-(1-{[3,5-dimethyl-7-(2-{[1-(methylsulfonyl)azetidin-3- yl]amino}ethoxy)tricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H-pyrazol-4-yl)pyridine-2- carboxylic acid; 3-{1-[(3-{2-[(3-amino-3-oxopropyl)amino]ethoxy}-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol-4-yl}-6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]pyridine-2-carboxylic acid; 6-[3-(1,3-benzothiazol-2- ylcarbamoyl)-1H-indazol-5-yl]-3-[1-({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec- 1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid; 6-[3-(1,3-benzothiazol-2- ylcarbamoyl)-1H-indol-5-yl]-3-[1-({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid; 6-[3-(1,3-benzothiazol-2- ylcarbamoyl)-1H-pyrrolo[2,3-b]pyridin-5-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid; 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-34-dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2- ((2-(N,N-dimethylsulfamoyl)ethyl)amino)ethoxy)-5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H- pyrazol-4-yl)picolinic acid; 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-3-{1-[(3-{2-[(3- hydroxypropyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol- 4-yl}pyridine-2-carboxylic acid; 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl]-3-(1-{[3-(2-{[3-(dimethylamino)-3-oxopropyl]amino}ethoxy)-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid; 6- [8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-(1-{[3,5-dimethyl-7-(2-{[3- (methylamino)-3-oxopropyl]amino}ethoxy)tricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H-pyrazol- 4-yl)pyridine-2-carboxylic acid; 3-(1-{[3-(2-aminoacetamido)-5,7-dimethyltricyclo[3.3.1.13,7]decan- 1-yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-{8-[(1,3-benzothiazol-2-yl)carbamoyl]-3,4- dihydroisoquinolin-2(1H)-yl}pyridine-2-carboxylic acid; 3-[1-({3-[(2-aminoethyl)sulfanyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]-6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]pyridine-2-carboxylic acid; 3-(1-{[3-(3-aminopropyl)- 5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]pyridine-2-carboxylic acid; and 3-(1-{[3-(2- aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]decan-1-yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-{5- [(1,3-benzothiazol-2-yl)carbamoyl]quinolin-3-yl}pyridine-2-carboxylic acid.
In one embodiment, the linker is cleavable by a lysosomal enzyme.
In one embodiment, the lysosomal enzyme is Cathepsin B.
In one embodiment, the linker comprises a segment according to structural formula (IVa), (IVb), (IVc), or (IVd):
Figure imgf000018_0001
Figure imgf000018_0002
Figure imgf000019_0001
Figure imgf000019_0002
wherein:peptide represents a peptide (illustrated N→C, wherein peptide includes the amino and carboxy“termini”) a cleavable by a lysosomal enzyme; T represents a polymer comprising one or more ethylene glycol units or an alkylene chain, or combinations thereof; Ra is selected from hydrogen, C1-6 alkyl, SO3H and CH2SO3H; Ry is hydrogen or C1-4 alkyl-(O)r-(C1-4 alkylene)s-G1 or C1-4 alkyl-(N)-[(C1-4 alkylene)-G1]2; Rz is C1-4 alkyl-(O)r-(C1-4 alkylene)s-G2; G1 is SO3H, CO2H, PEG 4- 32, or sugar moiety; G2 is SO3H, CO2H, or PEG 4-32 moiety; r is 0 or 1; s is 0 or 1; p is an integer ranging from 0 to 5; q is 0 or 1; x is 0 or 1; y is 0 or 1; represents the point of attachment of the linker to the Bcl-xL inhibitor; and * represents the point of attachment to the remainder of the linker.
In one embodiment, the peptide is selected from the group consisting of Val-Cit; Cit-Val; Ala-Ala; Ala-Cit; Cit-Ala; Asn-Cit; Cit-Asn; Cit-Cit; Val-Glu; Glu-Val; Ser-Cit; Cit-Ser; Lys-Cit; Cit-Lys; Asp-Cit; Cit-Asp; Ala-Val; Val-Ala; Phe-Lys; Lys-Phe; Val-Lys; Lys-Val; Ala-Lys; Lys- Ala; Phe-Cit; Cit-Phe; Leu-Cit; Cit-Leu; Ile-Cit; Cit-Ile; Phe-Arg; Arg-Phe; Cit-Trp; and Trp-Cit.
In one embodiment, the lysosomal enzyme is β-glucuronidase or β-galactosidase.
In one embodiment, the linker comprises a segment according to structural formula (Va), (Vb), (Vc), (Vd), or (Ve):
Figure imgf000019_0003
Figure imgf000020_0001
Figure imgf000020_0002
Figure imgf000020_0003
Figure imgf000020_0004
wherein q is 0 or 1; r is 0 or 1; X1 is CH2, O or NH; represents the point of attachment of the linker to the drug; and * represents the point of attachment to the remainder of the linker. In one embodiment, the linker comprises a segment according to structural formula (VIIIa), (VIIIb), or (VIIIc):
Figure imgf000021_0001
or a hydrolyzed derivative thereof, wherein Rq is H or–O-(CH2CH2O)11-CH3; x is 0 or 1; y is 0 or 1; G3 is–CH2CH2CH2SO3H or–CH2CH2O-(CH2CH2O)11-CH3; Rw is–O-CH2CH2SO3H or– NH(CO)-CH2CH2O-(CH2CH2O)12-CH3; * represents the point of attachment to the remainder of the linker; and represents the point of attachment of the linker to the antibody, wherein when in the hydrolyzed form, can be either at the α-position or β-position of the carboxylic acid next to it.
In one embodiment, the linker comprises a polyethylene glycol segment having from 1 to 6 ethylene glycol units.
In one embodiment, m is 2, 3 or 4.
In one embodiment, the linker L comprises a segment according to structural formula (IVa) or (IVb).
In one embodiment, the linker L is selected from the group consisting of IVa.1-IVa.8, IVb.1- IVb.19, IVc.1-IVc.7, IVd.1-IVd.4, Va.1-Va.12, Vb.1-Vb.10, Vc.1-Vc.11, Vd.1-Vd.6, Ve.1-Ve.2, VIa.1, VIc.1-V1c.2, VId.1-VId.4, VIIa.1-VIIa.4, VIIb.1-VIIb.8, VIIc.1-VIIc.6 in either the closed or open form.
In one embodiment, the linker L is selected from the group consisting of IVb.2, IVc.5, IVc.6, IVc.7, IVd.4, Vb.9, Vc.11, VIIa.1, VIIa.3, VIIc.1, VIIc.4, and VIIc.5, wherein the maleimide of each linker has reacted with the antibody Ab, forming a covalent attachment as either a succinimide (closed form) or succinamide (open form).
In one embodiment, the linker L is selected from the group consisting of IVb.2, IVc.5, IVc.6, IVd.4, Vc.11, VIIa.1, VIIa.3, VIIc.1, VIIc.4, VIIc.5, wherein the maleimide of each linker has reacted with the antibody Ab, forming a covalent attachment as either a succinimide (closed form) or succinamide (open form).
In one embodiment, the linker L is selected from the group consisting of IVb.2, Vc.11, VIIa.3, IVc.6, and VIIc.1, wherein is the attachment point to drug D and @ is the attachment point to the LK, wherein when the linker is in the open form as shown below, @ can be either at the α-position or β-position of the carboxylic acid next to it:
,
,
Figure imgf000022_0001
Figure imgf000023_0001
Figure imgf000024_0001
Figure imgf000025_0001
. In one embodiment, LK is a linkage formed with an amino group on the anti-hB7-H3 antibody Ab.
In one embodiment, LK is an amide or a thiourea.
In one embodiment, LK is a linkage formed with a sulfhydryl group on the anti-hB7-H3 antibody Ab.
In one embodiment, LK is a thioether.
In one embodiment, LK is selected from the group consisting of amide, thiourea and thioether; and m is an integer ranging from 1 to 8.
In one embodiment, D is the Bcl-xL inhibitor as described herein (e.g., W3.01, W3.02, W3.03, W3.04, W3.05, W3.06, W3.07, W3.08, W3.09, W3.10, W3.11, W3.12, W3.13, W3.14, W3.15, W3.16, W3.17, W3.18, W3.19, W3.20, W3.21, W3.22, W3.23, W3.24, W3.25, W3.26, W3.27, W3.28, W3.29, W3.30, W3.31, W3.32, W3.33, W3.34, W3.35, W3.36, W3.37, W3.38, W3.39, W3.40, W3.41, W3.42, W3.43, and pharmaceutically acceptable salts thereof); L is selected from the group consisting of linkers IVa.1-IVa.8, IVb.1-IVb.19, IVc.1-IVc.7, IVd.1-IVd.4, Va.1- Va.12, Vb.1-Vb.10, Vc.1-Vc.11, Vd.1-Vd.6, Ve.1-Ve.2, VIa.1, VIc.1-V1c.2, VId.1-VId.4, VIIa.1- VIIa.4, VIIb.1-VIIb.8, and VIIc.1-VIIc.6, wherein each linker has reacted with the antibody, Ab, forming a covalent attachment; LK is thioether; and m is an integer ranging from 1 to 8.
In one embodiment, D is the Bcl-xL inhibitor selected from the group consisting of the following compounds modified in that the hydrogen corresponding to the # position of structural formula (IIa) or (IIb) is not present, forming a monoradical: 3-(1-{[3-(2-aminoethoxy)-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[1-(1,3-benzothiazol-2- ylcarbamoyl)-5,6-dihydroimidazo[1,5-a]pyrazin-7(8H)-yl]pyridine-2-carboxylic acid; 6-[8-(1,3- benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid; 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]- 3-{1-[(3-{2-[(2-methoxyethyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5- methyl-1H-pyrazol-4-yl}pyridine-2-carboxylic acid; 3-(1-{[3-(2-aminoethoxy)-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-5-cyano-3,4-dihydroisoquinolin-2(1H)-yl]pyridine-2-carboxylic acid; 6-[4-(1,3- benzothiazol-2-ylcarbamoyl)isoquinolin-6-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid; and 3-{1-[(3-{2-[(3-amino-3-oxopropyl)amino]ethoxy}-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol-4-yl}-6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]pyridine-2-carboxylic acid; L is selected from the group consisting of linkers IVb.2, IVc.5, IVc.6, IVc.7, IVd.4, Vb.9, Vc.11, VIIa.1, VIIa.3, VIIc.1, VIIc.4, and VIIc.5 in either closed or open forms; LK is thioether; and m is an integer ranging from 2 to 4.
In one embodiment, the ADC is selected from the group consisting of huAb13v1-ZT, huAb13v1-ZZ, huAb13v1-SE, huAb13v1-SR, huAb3v2.5-ZT, huAb3v2.5-ZZ, huAb3v2.5-SE, huAb3v2.5-SR, huAb3v2.6-ZT, huAb3v2.6-ZZ, huAb3v2.6-SE, and huAb3v2.6-SR, wherein huAb13v1, huAb3v2.5, and huAb3v2.6 are the anti-hB7-H3 antibodies and KZ, SR, SE, XW, YG, ZT and ZZ are synthons disclosed in Table B, and wherein the conjugated synthons are either in open or closed form.
i iii
Figure imgf000026_0001
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
x v wherein m is an integer from 1 to 6. In one embodiment, Ab is an anti-hB7-H3 antibody, wherein the anti-hB7-H3 antibody comprises a heavy chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 35, a heavy chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 34, and a heavy chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 33; and a light chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 39, a light chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 38, and a light chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 37. In one embodiment, the Ab is an anti-hB7-H3 antibody, wherein the anti-hB7H3 antibody comprises a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 147, and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 144. In one embodiment, Ab is an anti-hB7-H3 antibody, wherein the anti-hB7-H3 antibody comprises a heavy chain constant region comprising the amino acid sequence set forth in SEQ ID NO: 160 and/or a light chain constant region comprising the amino acid sequence set forth in SEQ ID NO: 161. In one embodiment, Ab is an anti-hB7-H3 antibody, wherein the anti-hB7-H3 antibody comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 168, and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 169. In one embodiment, Ab is an anti-hB7-H3 antibody, wherein the anti-hB7-H3 antibody comprises a heavy chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 12, a heavy chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 140, and a heavy chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 10; and a light chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 15, a light chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 7, and a light chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 136. In one
embodiment, the Ab is an anti-hB7-H3 antibody, wherein the anti-hB7H3 antibody comprises a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 139, and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 135. In one embodiment, Ab is an anti-hB7-H3 antibody, wherein the anti-hB7-H3 antibody comprises a heavy chain constant region comprising the amino acid sequence set forth in SEQ ID NO: 160 and/or a light chain constant region comprising the amino acid sequence set forth in SEQ ID NO: 161. In one embodiment, Ab is an anti-hB7-H3 antibody, wherein the anti-hB7-H3 antibody comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 170, and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 171.
In one embodiment, m is an integer from 2 to 6. In one embodiment, m is 2.
In one embodiment, the ADC comprises an anti-hB7-H3 antibody comprising a heavy chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 12, a heavy chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 140, and a heavy chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 10; a light chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 15, a light chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 7, and a light chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 136 or 138.
In one embodiment, the ADC comprises an antibody comprising a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 139, and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 135.
In one embodiment, the ADC comprises an antibody comprising a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 139, and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 137.
In one embodiment, the ADC comprises an antibody comprising a light chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 39, a light chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 38, and a light chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 37; and a heavy chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 35 a heavy chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 34, and a heavy chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 33.
In one embodiment, the ADC comprises an antibody comprising a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 147, and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 144.
In one embodiment, the ADC is selected from the group consisting of huAb3v2.5-ZT, huAb3v2.5-ZZ, huAb3v2.5-XW, huAb3v2.5-SE, huAb3v2.5-SR, huAb3v2.5-YG, huAb3v2.5-KZ, huAb3v2.6-ZT, huAb3v2.6-ZZ, huAb3v2.6-XW, huAb3v2.6-SE, huAb3v2.6-SR, huAb3v2.6-YG, huAb3v2.6-KZ, huAb13v1-ZT, huAb13v1-ZZ, huAb13v1-XW, huAb13v1-SE, huAb13v1-SR, huAb13v1-YG, and huAb13v1-KZ.
In one aspect, the present invention provides a pharmaceutical composition comprising an effective amount of an ADC described herein, and a pharmaceutically acceptable carrier.
In one embodiment, the pharmaceutical composition comprises an ADC mixture comprising a plurality of the ADCs described herein, and a pharmaceutically acceptable carrier.
In one embodiment, the pharmaceutical composition comprises an ADC mixture having an average drug to antibody ratio (DAR) of 1.5 to 4.
In one embodiment, the pharmaceutical composition comprises an ADC mixture comprising ADCs each having a DAR of 1.5 to 8.
In one aspect, the present invention provides a method for treating cancer, comprising administering a therapeutically effective amount of the ADC described herein to a subject in need thereof.
In one embodiment, the cancer is selected from the group consisting of small cell lung cancer, non small cell lung cancer, breast cancer, ovarian cancer, a glioblastoma, prostate cancer, pancreatic cancer, colon cancer, gastric cancer, melanoma, hepatocellular carcinoma, head and neck cancer, kidney cancer, leukemia, e.g., acute myeloid leukemia (AML), and lymphoma, e.g., non-Hodgkin's lymphoma (NHL). In one embodiment, the cancer is a squamous cell carcinoma. In one
embodiment, the squamous cell carcinoma is squamous lung cancer or squamous head and neck cancer. In one embodiment, the cancer is triple negative breast cancer. In one embodiment, the cancer is non-small cell lung cancer.
In one aspect, the present invention provides a method for inhibiting or decreasing solid tumor growth in a subject having a solid tumor, said method comprising administering an effective amount of the ADC described herein to the subject having the solid tumor, such that the solid tumor growth is inhibited or decreased.
In one embodiment, the solid tumor is a non-small cell lung carcinoma.
In one embodiment, the cancer is characterized as having an activating EGFR mutation. In one embodiment, the activating EGFR mutation is selected from the group consisting of an exon 19 deletion mutation, a single-point substitution mutation L858R in exon 21, a T790M point mutation, and combinations thereof.
In one embodiment, the ADC is administered in combination with an additional agent or an additional therapy. In one embodiment, the additional agent is selected from the group consisting of an anti-PD1 antibody (e.g. pembrolizumab), an anti-PD-L1 antibody (e.g. atezolizumab), an anti- CTLA-4 antibody (e.g. ipilimumab), a MEK inhibitor (e.g. trametinib), an ERK inhibitor, a BRAF inhibitor (e.g. dabrafenib), osimertinib, erlotinib, gefitinib, sorafenib, a CDK9 inhibitor (e.g.
dinaciclib), a MCL-1 inhibitor, temozolomide, a Bcl-xL inhibitor, a Bcl-2 inhibitor (e.g. venetoclax), ibrutinib, a mTOR inhibitor (e.g. everolimus), a PI3K inhibitor (e.g. buparlisib), duvelisib, idelalisib, an AKT inhibitor, a HER2 inhibitor (e.g. lapatinib), a taxane (e.g. docetaxel, paclitaxel, nab- paclitaxel), an ADC comprising an auristatin, an ADC comprising a PBD (e.g. rovalpituzumab tesirine), an ADC comprising a maytansinoid (e.g. TDM1), a TRAIL agonist, a proteasome inhibitor (e.g. bortezomib), and a nicotinamide phosphoribosyltransferase (NAMPT) inhibitor.
In one embodiment, the additional therapy is radiation. In another embodiment, the additional agent is a chemotherapeutic agent.
In one embodiment, the anti-B7-H3 ADCs of the invention are administered in combination with venetoclax to a human subject for the treatment of small cell lung cancer (SCLC).
In one aspect, the present invention provides a process for the preparation of an ADC according to structural formula (I):
Figure imgf000035_0001
wherein:
D is the Bcl-xL inhibitor drug of formula (IIa) or (IIb) as disclosed herein;
L is the linker as disclosed herein;
Ab is an hB7-H3 antibody, wherein the hB7-H3 antibody comprises the heavy and light chain CDRs of huAb3v2.5, huAb3v2.6, or huAb13v1;
LK represents a covalent linkage linking linker L to antibody Ab; and
m is an integer ranging from 1 to 20;
the process comprising:
treating an antibody in an aqueous solution with an effective amount of a disulfide reducing agent at 30-40 °C for at least 15 minutes, and then cooling the antibody solution to 20-27 °C;
adding to the reduced antibody solution a solution of water/dimethyl sulfoxide comprising a synthon selected from the group of 2.1 to 2.31 and 2.34 to 2.72 (Table B);
adjusting the pH of the solution to a pH of 7.5 to 8.5;
allowing the reaction to run for 48 to 80 hours to form the ADC; wherein the mass is shifted by 18 ± 2 amu for each hydrolysis of a succinimide to a succinamide as measured by electron spray mass spectrometry; and
wherein the ADC is optionally purified by hydrophobic interaction chromatography.
In one embodiment, m is 2.
In another aspect, the present invention provides an ADC prepared by the process as described above. BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is a graphical representation of the epitope grouping of murine anti-B7-H3 hybridoma antibodies as determined by pair-wise binding assays.
Figure 2 depicts an antibody reduction, modification with a maleimide derivative to give a thiosuccinimide intermediate, and subsequent hydrolysis of thiosuccinimide moiety
Figure 3 depicts the structure of an antibody-maleimidocaproyl-vc-PABA-MMAE ADC. Figure 4 depicts the structure of a PBD dimer (SGD-1882) conjugated to an antibody (Ab) via a maleimidocaproyl-valine-alanine linker (collectively referred to as SGD-1910).
Figure 5 depicts the MS characterization of light chain and heavy chain of huAb13v11) prior to conjugation, 2) after conjugation to a maleimide derivative to give a thiosuccinimide intermediate and 3) post pH 8-mediated hydrolysis of the thiosuccinimide ring. DETAILED DESCRIPTION OF THE INVENTION
Various aspects of the invention relate to anti-B7-H3 antibodies and antibody fragments, anti- B7-H3 ADCs, and pharmaceutical compositions thereof, as well as nucleic acids, recombinant expression vectors and host cells for making such antibodies and fragments. Methods of using the antibodies, fragments, and ADCs described herein to detect human B7-H3, to inhibit human B7-H3 activity (in vitro or in vivo), and to treat cancers are also encompassed by the invention. In certain embodiments, the invention provides anti-B7-H3 ADCs, including ADCs comprising Bcl-xL inhibitors, synthons useful for synthesizing the ADCs, compositions comprising the ADCs, methods of making the ADCs, and various methods of using the ADCs.
As will be appreciated by skilled artisans, the ADCs disclosed herein are“modular” in nature. Throughout the instant disclosure, various specific embodiments of the various“modules” comprising the ADCs, as well as the synthons useful for synthesizing the ADCs, are described. As specific non- limiting examples, specific embodiments of antibodies, linkers, and Bcl-xL inhibitors that may comprise the ADCs and synthons are described. It is intended that all of the specific embodiments described may be combined with each other as though each specific combination were explicitly described individually.
It will also be appreciated by skilled artisans that the various ADCs and/or ADC synthons described herein may be in the form of salts, and in certain embodiments, particularly pharmaceutically acceptable salts. The compounds of the present disclosure that possess a sufficiently acidic, a sufficiently basic, or both functional groups, can react with any of a number of inorganic bases, and inorganic and organic acids, to form a salt. Alternatively, compounds that are inherently charged, such as those with a quaternary nitrogen, can form a salt with an appropriate counterion, e.g., a halide such as a bromide, chloride, or fluoride.
Acids commonly employed to form acid addition salts are inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, phosphoric acid, and the like, and organic acids such as p-toluenesulfonic acid, methanesulfonic acid, oxalic acid, p-bromophenyl- sulfonic acid, carbonic acid, succinic acid, citric acid, etc. Base addition salts include those derived from inorganic bases, such as ammonium and alkali or alkaline earth metal hydroxides, carbonates, bicarbonates, and the like.
In the disclosure below, if both structural diagrams and nomenclature are included and if the nomenclature conflicts with the structural diagram, the structural diagram controls.
An outline of the Detailed Description of the Invention is provided below:
I. Definitions
II. Anti-B7-H3 Antibodies
II.A. Anti-B7-H3 Chimeric Antibodies
II.B. Humanized Anti-B7-H3 Antibodies
III. Anti-B7-H3 Antibody Drug Conjugates (ADCs)
III.A. Anti-B7-H3 / Bcl-xL Inhibitor ADCs
III.A.1. Bcl-xL Inhibitors
III.A.2. Bcl-xL Linkers
Cleavable Linkers
Non-Cleavable Linkers
Groups Used to Attach Linkers to Anti-B7-H3 Antibodies
Linker Selection Considerations
III.A.3. Bcl-xL ADC Synthons
III.A.4. Methods of Synthesis of Bcl-xL ADCs
III.A.5. General Methods for Synthesizing Bcl-xL Inhibitors III.A.6. General Methods for Synthesizing Synthons
III.A.7. General Methods for Synthesizing Anti-B7-H3 ADCs III.B. Anti-B7-H3 ADCs: Other Exemplary Drugs for Conjugation
III.C. Anti-B7-H3 ADCs: Other Exemplary Linkers
IV. Purification of Anti-B7-H3 ADCs
V. Uses of Anti-B7-H3 Antibodies and Anti-B7-H3 ADCs
VI. Pharmaceutical Compositions I. Definitions
In order that the invention may be more readily understood, certain terms are first defined. In addition, it should be noted that whenever a value or range of values of a parameter are recited, it is intended that values and ranges intermediate to the recited values are also intended to be part of this invention.
The term“anti-B7-H3 antibody” refers to an antibody that specifically binds to B7-H3. An antibody“which binds” an antigen of interest, i.e., B7-H3, is one capable of binding that antigen with sufficient affinity such that the antibody is useful in targeting a cell expressing the antigen. In a preferred embodiment, the antibody specifically binds to human B7-H3 (hB7-H3). Examples of anti- B7-H3 antibodies are disclosed in the examples below. Unless otherwise indicated, the term“anti- B7-H3 antibody” is meant to refer to an antibody which binds to wild type B7-H3 (e.g., a 4IgB7-H3 isoform of B7-H3) or any variant of B7-H3. The amino acid sequence of wild type human B7-H3 is provided below as SEQ ID NO: 149, where the signal peptide (amino acid residues 1-28) is underlined.
MLRRRGSPGMGVHVGAALGALWFCLTGALEVQVPEDPVVALVGTDATLCCSFSPEPGFSLAQLNLIWQ LTDTKQLVHSFAEGQDQGSAYANRTALFPDLLAQGNASLRLQRVRVADEGSFTCFVSIRDFGSAAVSL QVAAPYSKPSMTLEPNKDLRPGDTVTITCSSYQGYPEAEVFWQDGQGVPLTGNVTTSQMANEQGLFDV HSILRVVLGANGTYSCLVRNPVLQQDAHSSVTITPQRSPTGAVEVQVPEDPVVALVGTDATLRCSFSP EPGFSLAQLNLIWQLTDTKQLVHSFTEGRDQGSAYANRTALFPDLLAQGNASLRLQRVRVADEGSFTC FVSIRDFGSAAVSLQVAAPYSKPSMTLEPNKDLRPGDTVTITCSSYRGYPEAEVFWQDGQGVPLTGNV TTSQMANEQGLFDVHSVLRVVLGANGTYSCLVRNPVLQQDAHGSVTITGQPMTFPPEALWVTVGLSVC LIALLVALAFVCWRKIKQSCEEENAGAEDQDGEGEGSKTALQPLKHSDSKEDDGQEIA (SEQ ID NO: 149) Thus, in one embodiment of the invention, the antibody or ADC binds human B7-H3 as defined in SEQ ID NO: 149. The extracellular domain (ECD) of human B7-H3 is provided in SEQ ID NO: 152 (inclusive of a His tag). As such, in one embodiment of the invention, the antibody of ADC binds the ECD of human B7-H3 as described in the ECD of SEQ ID NO: 152.
The terms“specific binding” or“specifically binding”, as used herein, in reference to the interaction of an antibody or an ADC with a second chemical species, mean that the interaction is dependent upon the presence of a particular structure (e.g., an antigenic determinant or epitope) on the chemical species; for example, an antibody recognizes and binds to a specific protein structure rather than to proteins generally. If an antibody or ADC is specific for epitope“A”, the presence of a molecule containing epitope A (or free, unlabeled A), in a reaction containing labeled“A” and the antibody, will reduce the amount of labeled A bound to the antibody or ADC. By way of example, an antibody "binds specifically" to a target if the antibody, when labeled, can be competed away from its target by the corresponding non-labeled antibody. In one embodiment, an antibody specifically binds to a target, e.g., B7-H3, if the antibody has a KD for the target of at least about 10-4 M, 10-5 M, 10-6 M, 10-7 M, 10-8 M, 10-9 M, 10-10 M, 10-11 M, 10-12 M, or less (less meaning a number that is less than 10- 12 , e.g.10-13). In one embodiment, the term“specific binding to B7-H3” or“specifically binds to B7- H3,” as used herein, refers to an antibody or an ADC that binds to B7-H3 and has a dissociation constant (KD) of 1.0 x 10-7 M or less, as determined by surface plasmon resonance. It shall be understood, however, that the antibody or ADC may be capable of specifically binding to two or more antigens which are related in sequence. For example, in one embodiment, an antibody can specifically bind to both human and a non-human (e.g., mouse or non-human primate) orthologs of B7-H3.
The term“antibody” or“Ab” refers to an immunoglobulin molecule that specifically binds to an antigen and comprises a heavy (H) chain(s) and a light (L chain(s). Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CH1, CH2 and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino- terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. An antibody can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY) and class (e.g., IgG1, IgG2, IgG 3, IgG4, IgA1 and IgA2) or subclass. While the term“antibody” is not intended to include antigen binding portions of an antibody (defined below), it is intended, in certain embodiments, to describe an antibody comprising a small number of amino acid deletions from the carboxy end of the heavy chain(s). Thus, in one embodiment, an antibody comprises a heavy chain having 1-5 amino acid deletions the carboxy end of the heavy chain. In one embodiment, an antibody is a monoclonal antibody which is an IgG, having four polypeptide chains, two heavy (H) chains, and two light (L chains) that can bind to hB7-H3. In one embodiment, an antibody is a monoclonal IgG antibody comprising a lambda or a kappa light chain.
The term“antigen binding portion” or“antigen binding fragment” of an antibody (or simply “antibody portion” or“antibody fragment”), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., hB7-H3). It has been shown that the antigen binding function of an antibody can be performed by fragments of a full-length antibody. Such antibody embodiments may also be bispecific, dual specific, or multi-specific formats; specifically binding to two or more different antigens. Examples of binding fragments encompassed within the term“antigen binding portion” of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341:544- 546, Winter et al., PCT publication WO 90/05144 A1 herein incorporated by reference), which comprises a single variable domain; and (vi) an isolated complementarity determining region (CDR). Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883). Such single chain antibodies are also intended to be encompassed within the term“antigen binding portion” of an antibody. In certain embodiments of the invention, scFv molecules may be incorporated into a fusion protein. Other forms of single chain antibodies, such as diabodies are also encompassed. Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R.J., et al. (1994) Structure 2:1121-1123). Such antibody binding portions are known in the art (Kontermann and Dubel eds., Antibody Engineering (2001) Springer-Verlag. New York.790 pp. (ISBN 3-540-41354-5).
An IgG (Immunoglobuin G) is a class of antibody comprising two heavy chains and two light chains arranged in a Y-shape. Exemplary human IgG heavy chain and light chain constant domain amino acid sequences are known in the art and represented below in Table A. Table A: Sequences of human IgG heavy chain constant domains and light chain constant domains
Figure imgf000040_0001
Figure imgf000041_0001
An“isolated antibody”, as used herein, is intended to refer to an antibody that is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds B7-H3 is substantially free of antibodies that specifically bind antigens other than B7-H3). An isolated antibody that specifically binds B7-H3 may, however, have cross-reactivity to other antigens, such as B7-H3 molecules from other species. Moreover, an isolated antibody may be substantially free of other cellular material and/or chemicals.
The term“humanized antibody” refers to antibodies which comprise heavy and light chain variable region sequences from a nonhuman species (e.g., a mouse) but in which at least a portion of the VH and/or VL sequence has been altered to be more“human-like”, i.e., more similar to human germline variable sequences. In particular, the term“humanized antibody” is an antibody or a variant, derivative, analog or fragment thereof which immunospecifically binds to an antigen of interest and which comprises a framework (FR) region having substantially the amino acid sequence of a human antibody and a complementary determining region (CDR) having substantially the amino acid sequence of a non-human antibody. As used herein, the term“substantially” in the context of a CDR refers to a CDR having an amino acid sequence at least 80%, preferably at least 85%, at least 90%, at least 95%, at least 98% or at least 99% identical to the amino acid sequence of a non-human antibody CDR. A humanized antibody comprises substantially all of at least one, and typically two, variable domains (Fab, Fab', F(ab')2, FabC, Fv) in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin (i.e., donor antibody) and all or substantially all of the framework regions are those of a human immunoglobulin consensus sequence. Preferably, a humanized antibody also comprises at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. In some embodiments, a humanized antibody contains both the light chain as well as at least the variable domain of a heavy chain. The antibody also may include the CH1, hinge, CH2, CH3, and CH4 regions of the heavy chain. In some embodiments, a humanized antibody only contains a humanized light chain. In other embodiments, a humanized antibody only contains a humanized heavy chain. In specific embodiments, a humanized antibody only contains a humanized variable domain of a light chain and/or humanized heavy chain.
The humanized antibody can be selected from any class of immunoglobulins, including IgM, IgG IgD IgA and IgE and any isotype including without limitation IgG1 IgG2 IgG3 and IgG4 The humanized antibody may comprise sequences from more than one class or isotype, and particular constant domains may be selected to optimize desired effector functions using techniques well-known in the art.
The terms“Kabat numbering,”“Kabat definitions,” and“Kabat labeling” are used interchangeably herein. These terms, which are recognized in the art, refer to a system of numbering amino acid residues which are more variable (i.e., hypervariable) than other amino acid residues in the heavy and light chain variable regions of an antibody, or an antigen binding portion thereof (Kabat et al. (1971) Ann. NY Acad, Sci.190:382-391 and, Kabat, E.A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH
Publication No.91-3242). For the heavy chain variable region, the hypervariable region ranges from amino acid positions 31 to 35 for CDR1, amino acid positions 50 to 65 for CDR2, and amino acid positions 95 to 102 for CDR3. For the light chain variable region, the hypervariable region ranges from amino acid positions 24 to 34 for CDR1, amino acid positions 50 to 56 for CDR2, and amino acid positions 89 to 97 for CDR3.
As used herein, the term“CDR” refers to the complementarity determining region within antibody variable sequences. There are three CDRs in each of the variable regions of the heavy chain (HC) and the light chain (LC), which are designated CDR1, CDR2 and CDR3 (or specifically HC CDR1, HC CDR2, HC CDR3, LC CDR1, LC CDR2, and LC CDR3), for each of the variable regions. The term“CDR set” as used herein refers to a group of three CDRs that occur in a single variable region capable of binding the antigen. The exact boundaries of these CDRs have been defined differently according to different systems. The system described by Kabat (Kabat et al., Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md. (1987) and (1991)) not only provides an unambiguous residue numbering system applicable to any variable region of an antibody, but also provides precise residue boundaries defining the three CDRs. These CDRs may be referred to as Kabat CDRs. Chothia and coworkers (Chothia &Lesk, J. Mol. Biol.196:901-917 (1987) and Chothia et al., Nature 342:877-883 (1989)) found that certain sub- portions within Kabat CDRs adopt nearly identical peptide backbone conformations, despite having great diversity at the level of amino acid sequence. These sub-portions were designated as L1, L2 and L3 or H1, H2 and H3 where the“L” and the“H” designates the light chain and the heavy chains regions, respectively. These regions may be referred to as Chothia CDRs, which have boundaries that overlap with Kabat CDRs. Other boundaries defining CDRs overlapping with the Kabat CDRs have been described by Padlan (FASEB J.9:133-139 (1995)) and MacCallum (J. Mol. Biol.262(5):732-45 (1996)). Still other CDR boundary definitions may not strictly follow one of the above systems, but will nonetheless overlap with the Kabat CDRs, although they may be shortened or lengthened in light of prediction or experimental findings that particular residues or groups of residues or even entire CDRs do not significantly impact antigen binding. The methods used herein may utilize CDRs defined according to any of these systems although preferred embodiments use Kabat or Chothia defined CDRs As used herein, the term“framework” or“framework sequence” refers to the remaining sequences of a variable region minus the CDRs. Because the exact definition of a CDR sequence can be determined by different systems, the meaning of a framework sequence is subject to
correspondingly different interpretations. The six CDRs (CDR-L1, CDR-L2, and CDR-L3 of light chain and CDR-H1, CDR-H2, and CDR-H3 of heavy chain) also divide the framework regions on the light chain and the heavy chain into four sub-regions (FR1, FR2, FR3 and FR4) on each chain, in which CDR1 is positioned between FR1 and FR2, CDR2 between FR2 and FR3, and CDR3 between FR3 and FR4. Without specifying the particular sub-regions as FR1, FR2, FR3 or FR4, a framework region, as referred by others, represents the combined FR's within the variable region of a single, naturally occurring immunoglobulin chain. As used herein, a FR represents one of the four sub- regions, and FRs represents two or more of the four sub- regions constituting a framework region.
The framework and CDR regions of a humanized antibody need not correspond precisely to the parental sequences, e.g., the donor antibody CDR or the consensus framework may be mutagenized by substitution, insertion and/or deletion of at least one amino acid residue so that the CDR or framework residue at that site does not correspond to either the donor antibody or the consensus framework. In a preferred embodiment, such mutations, however, will not be extensive. Usually, at least 80%, preferably at least 85%, more preferably at least 90%, and most preferably at least 95% of the humanized antibody residues will correspond to those of the parental FR and CDR sequences. As used herein, the term“consensus framework” refers to the framework region in the consensus immunoglobulin sequence. As used herein, the term“consensus immunoglobulin sequence” refers to the sequence formed from the most frequently occurring amino acids (or nucleotides) in a family of related immunoglobulin sequences (See e.g., Winnaker, From Genes to Clones (Verlagsgesellschaft, Weinheim, Germany 1987). In a family of immunoglobulins, each position in the consensus sequence is occupied by the amino acid occurring most frequently at that position in the family. If two amino acids occur equally frequently, either can be included in the consensus sequence.
The term“human acceptor framework”, as used herein, is meant to refer to a framework of an antibody or antibody fragment thereof comprising the amino acid sequence of a VH or VL framework derived from a human antibody or antibody fragment thereof or a human consensus sequence framework into which CDRs from a non-human species may be incorporated.
“Percent (%) amino acid sequence identity” with respect to a peptide or polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the specific peptide or polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art for instance using publicly available computer software such as BLAST BLAST-2 ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. In one embodiment, the invention includes an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identity to an amino acid sequence set forth in any one of SEQ ID NOs: 1 to 148.
The term“multivalent antibody” is used herein to denote an antibody comprising two or more antigen binding sites. In certain embodiments, the multivalent antibody may be engineered to have the three or more antigen binding sites, and is generally not a naturally occurring antibody.
The term“multispecific antibody” refers to an antibody capable of binding two or more unrelated antigens. In one embodiment, the multispecific antibody is a bispecific antibody that is capable of binding to two unrelated antigens, e.g., a bispecific antibody, or antigen-binding portion thereof, that binds B7-H3 and CD3.
The term“dual variable domain” or“DVD,” as used interchangeably herein, are antigen binding proteins that comprise two or more antigen binding sites and are tetravalent or multivalent binding proteins. Such DVDs may be monospecific, i.e., capable of binding one antigen or multispecific, i.e. capable of binding two or more antigens. DVD binding proteins comprising two heavy chain DVD polypeptides and two light chain DVD polypeptides are referred to a DVD Ig. Each half of a DVD Ig comprises a heavy chain DVD polypeptide, and a light chain DVD
polypeptide, and two antigen binding sites. Each binding site comprises a heavy chain variable domain and a light chain variable domain with a total of 6 CDRs involved in antigen binding per antigen binding site. In one embodiment, the CDRs described herein are used in an anti-B7-H3 DVD.
The term "chimeric antigen receptor" or "CAR" refers to a recombinant protein comprising at least (1) an antigen-binding region, e.g., a variable heavy or light chain of an antibody, (2) a transmembrane domain to anchor the CAR into a T cell, and (3) one or more intracellular signaling domains.
The term“activity” includes activities such as the binding specificity/affinity of an antibody or ADC for an antigen, for example, an anti-hB7-H3 antibody that binds to an hB7-H3 antigen and/or the neutralizing potency of an antibody, for example, an anti-hB7-H3 antibody whose binding to hB7- H3 inhibits the biological activity of hB7-H3, e.g., inhibition of proliferation of B7-H3 expressing cell lines, e.g., human H146 lung carcinoma cells, human H1650 lung carcinoma cells, or human EBC1 lung carcinoma cells.
The term“non small-cell lung carcinoma (NSCLC) xenograft assay,” as used herein, refers to an in vivo assay used to determine whether an anti-B7-H3 antibody or ADC, can inhibit tumor growth (e.g., further growth) and/or decrease tumor growth resulting from the transplantation of NSCLC cells into an immunodeficient mouse. An NSCLC xenograft assay includes transplantation of NSCLC cells into an immunodeficient mouse such that a tumor grows to a desired size e g 200-250 mm3 whereupon the antibody or ADC is administered to the mouse to determine whether the antibody or ADC can inhibit and/or decrease tumor growth. In certain embodiments, the activity of the antibody or ADC is determined according to the percent tumor growth inhibition (%TGI) relative to a control antibody, e.g., a human IgG antibody (or collection thereof) which does not specifically bind tumor cells, e.g., is directed to an antigen not associated with cancer or is obtained from a source which is noncancerous (e.g., normal human serum). In such embodiments, the antibody (or ADC) and the control antibody are administered to the mouse at the same dose, with the same frequency, and via the same route. In one embodiment, the mouse used in the NSCLC xenograft assay is a severe combined immunodeficiency (SCID) mouse and/or an athymic CD-1 nude mouse. Examples of NSCLC cells that may be used in the NSCLC xenograft assay include, but are not limited to, H1299 cells (NCI- H1299 [H-1299] (ATCC® CRL-5803)), H1650 cells (NCI-H1650 [H-1650] (ATCC® CRL-5883™)), H1975 cells (NCI-H1975 cells [H1975] (ATCC® CRL-5908™)), and EBC-1 cells.
The term“small-cell lung carcinoma (SCLC) xenograft assay,” as used herein, refers to an in vivo assay used to determine whether an anti-B7-H3 antibody or ADC, can inhibit tumor growth (e.g., further growth) and/or decrease tumor growth resulting from the transplantation of SCLC cells into an immunodeficient mouse. An SCLC xenograft assay includes transplantation of SCLC cells into an immunodeficient mouse such that a tumor grows to a desired size, e.g., 200-250 mm3, whereupon the antibody or ADC is administered to the mouse to determine whether the antibody or ADC can inhibit and/or decrease tumor growth. In certain embodiments, the activity of the antibody or ADC is determined according to the percent tumor growth inhibition (%TGI) relative to a control antibody, e.g., a human IgG antibody (or collection thereof) which does not specifically bind tumor cells, e.g., is directed to an antigen not associated with cancer or is obtained from a source which is noncancerous (e.g., normal human serum). In such embodiments, the antibody (or ADC) and the control antibody are administered to the mouse at the same dose, with the same frequency, and via the same route. In one embodiment, the mouse used in the NSCLC xenograft assay is a severe combined
immunodeficiency (SCID) mouse and/or an athymic CD-1 nude mouse. Examples of SCLC cells that may be used in the SCLC xenograft assay include, but are not limited to, H146 cells (NCI-H146 cells [H146] (ATCC® HTB-173™)), and H847 cells (NCI-H847 [H847] (ATCC® CRL-5846™)).
The term“epitope” refers to a region of an antigen that is bound by an antibody or ADC. In certain embodiments, epitope determinants include chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl, or sulfonyl, and, in certain embodiments, may have specific three dimensional structural characteristics, and/or specific charge characteristics. In certain embodiments, an antibody is said to specifically bind an antigen when it preferentially recognizes its target antigen in a complex mixture of proteins and/or macromolecules.
The term“surface plasmon resonance”, as used herein, refers to an optical phenomenon that allows for the analysis of real-time biospecific interactions by detection of alterations in protein concentrations within a biosensor matrix for example using the BIAcore system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, NJ). For further descriptions, see Jönsson, U., et al. (1993) Ann. Biol. Clin.51:19-26; Jönsson, U., et al. (1991) Biotechniques 11:620-627; Johnsson, B., et al. (1995) J. Mol. Recognit.8:125-131; and Johnnson, B., et al. (1991) Anal. Biochem.198:268- 277. In one embodiment, surface plasmon resonance is determined according to the methods described in Example 2.
The term“ kon” or“ ka”, as used herein, is intended to refer to the on rate constant for association of an antibody to the antigen to form the antibody/antigen complex.
The term“koff” or“ kd”, as used herein, is intended to refer to the off rate constant for dissociation of an antibody from the antibody/antigen complex.
The term“KD”, as used herein, is intended to refer to the equilibrium dissociation constant of a particular antibody-antigen interaction (e.g., huAb13 antibody and B7-H3). KD is calculated by ka / kd.
The term“competitive binding”, as used herein, refers to a situation in which a first antibody competes with a second antibody, for a binding site on a third molecule, e.g., an antigen. In one embodiment, competitive binding between two antibodies is determined using FACS analysis.
The term“competitive binding assay” is an assay used to determine whether two or more antibodies bind to the same epitope. In one embodiment, a competitive binding assay is a competition fluorescent activated cell sorting (FACS) assay which is used to determine whether two or more antibodies bind to the same epitope by determining whether the fluorescent signal of a labeled antibody is reduced due to the introduction of a non-labeled antibody, where competition for the same epitope will lower the level of fluorescence.
The term“labeled antibody” as used herein, refers to an antibody, or an antigen binding portion thereof, with a label incorporated that provides for the identification of the binding protein, e.g., an antibody. Preferably, the label is a detectable marker, e.g., incorporation of a radiolabeled amino acid or attachment to a polypeptide of biotinyl moieties that can be detected by marked avidin (e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or colorimetric methods). Examples of labels for polypeptides include, but are not limited to, the following: radioisotopes or radionuclides (e.g., 3H 14
, C 35
, S, 90Y, 99Tc, 111In, 125I, 131I, 177Lu, 166Ho, or 153Sm); fluorescent labels (e.g., FITC, rhodamine, lanthanide phosphors), enzymatic labels (e.g., horseradish peroxidase, luciferase, alkaline phosphatase); chemiluminescent markers; biotinyl groups; predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags); and magnetic agents, such as gadolinium chelates.
The term“antibody-drug-conjugate” or“ADC” refers to a binding protein, such as an antibody or antigen binding fragment thereof, chemically linked to one or more chemical drug(s) (also referred to herein as agent(s), warhead(s), or payload(s)) that may optionally be therapeutic or cytotoxic agents In a preferred embodiment an ADC includes an antibody a drug (e g a cytotoxic drug), and a linker that enables attachment or conjugation of the drug to the antibody. An ADC typically has anywhere from 1 to 8 drugs conjugated to the antibody, including drug loaded species of 2, 4, 6, or 8. Non-limiting examples of drugs that may be included in the ADCs are mitotic inhibitors, antitumor antibiotics, immunomodulating agents, vectors for gene therapy, alkylating agents, antiangiogenic agents, antimetabolites, boron-containing agents, chemoprotective agents, hormones, antihormone agents, corticosteroids, photoactive therapeutic agents, oligonucleotides, radionuclide agents, topoisomerase inhibitors, kinase inhibitors (e.g., TEC-family kinase inhibitors and serine/threonine kinase inhibitors), and radiosensitizers. In one embodiment, the drug is a Bcl-xL inhibitor.
The terms“anti-B7-H3 antibody drug conjugate” or“anti-B7-H3 ADC”, used
interchangeably herein, refer to an ADC comprising an antibody that specifically binds to B7-H3, whereby the antibody is conjugated to one or more chemical agent(s). In one embodiment, the anti- B7-H3 ADC comprises antibody huAb13v1, huAb3v2.5, or huAb3v2.6 conjugated to an auristatin, e.g., MMAE or MMAF. In one embodiment, the anti-B7-H3 ADC comprises antibody huAb13v1, huAb3v2.5, or huAb3v2.6 conjugated to a Bcl-xL inhibitor. In a preferred embodiment, the anti-B7- H3B7-H3 ADC binds to human B7-H3B7-H3B7-H3.
The term“Bcl-xL inhibitor”, as used herein, refers to a compound which antagonizes Bcl-xL activity in a cell. In one embodiment, a Bcl-xL inhibitor promotes apoptosis of a cell by inhibiting Bcl-xL activity.
The term“auristatin”, as used herein, refers to a family of antimitotic agents. Auristatin derivatives are also included within the definition of the term“auristatin”. Examples of auristatins include, but are not limited to, auristatin E (AE), monomethylauristatin E (MMAE),
monomethylauristatin F (MMAF), and synthetic analogs of dolastatin. In one embodiment, an anti- B7-H3 antibody described herein is conjugated to an auristatin to form an anti-B7-H3 ADC.
As used herein, the term“Ab-vcMMAE” is used to refer to an ADC comprising an antibody conjugated to monomethylauristatin E (MMAE) via a maleimidocaproyl valine citrulline p- aminobenzyloxycarbamyl (PABA) linker.
As used herein , the term“mcMMAF” is used to refer to a linker/drug combination of maleimidocaproyl-monomethylauristatin F (MMAF).
The term“drug-to-antibody ratio” or“DAR” refers to the number of drugs, e.g., a Bcl-xL inhibitor, attached to the antibody of the ADC. The DAR of an ADC can range from 1 to 8, although higher loads, e.g., 20, are also possible depending on the number of linkage site on an antibody. The term DAR may be used in reference to the number of drugs loaded onto an individual antibody, or, alternatively, may be used in reference to the average or mean DAR of a group of ADCs.
The term“undesired ADC species”, as used herein, refers to any drug loaded species which is to be separated from an ADC species having a different drug load. In one embodiment, the term undesired ADC species may refer to drug loaded species of 6 or more i e ADCs with a DAR of 6 or more, including DAR6, DAR7, DAR8, and DAR greater than 8 (i.e., drug loaded species of 6, 7, 8, or greater than 8). In a separate embodiment, the term undesired ADC species may refer to drug loaded species of 8 or more, i.e., ADCs with a DAR of 8 or more, including DAR8, and DAR greater than 8 (i.e., drug loaded species of 8, or greater than 8).
The term“ADC mixture”, as used herein, refers to a composition containing a heterogeneous DAR distribution of ADCs. In one embodiment, an ADC mixture contains ADCs having a distribution of DARs of 1 to 8, e.g., 1.5, 2, 4, 6, and 8 (i.e., drug loaded species of 1.5, 2, 4, 6, and 8). Notably, degradation products may result such that DARs of 1, 3, 5, and 7 may also be included in the mixture. Further, ADCs within the mixture may also have DARs greater than 8. The ADC mixture results from interchain disulfide reduction followed by conjugation. In one embodiment, the ADC mixture comprises both ADCs with a DAR of 4 or less (i.e., a drug loaded species of 4 or less) and ADCs with a DAR of 6 or more (i.e., a drug loaded species of 6 or more).
The term a“xenograft assay”, as used herein, refers to a human tumor xenograft assay, wherein human tumor cells are transplanted, either under the skin or into the organ type in which the tumor originated, into immunocompromised mice that do not reject human cells.
The term“cancer” is meant to refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. Examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies. More particular examples of such cancers include glioblastoma, acute myeloid leukemia (AML), non- Hodgkin's lymphoma (NHL), non-small cell lung cancer, lung cancer, colon cancer, colorectal cancer, head and neck cancer, breast cancer (e.g., triple negative breast cancer), pancreatic cancer, squamous cell tumors, squamous cell carcinoma (e.g., squamous cell lung cancer or squamous cell head and neck cancer), anal cancer, skin cancer, and vulvar cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having a tumor(s) that overexpresses B7-H3. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having a solid tumor which is likely to overexpress B7-H3. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having squamous cell non-small cell lung cancer (NSCLC). In one embodiment, the antibodies or ADCs of the invention are administered to a patient having solid tumors, including advanced solid tumors. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having prostate cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having non-small cell lung cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having a glioblastoma. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having colon cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having head and neck cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having kidney cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having clear cell renal cell carcinoma. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having glioma. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having melanoma. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having pancreatic cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having gastric cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having ovarian cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having colorectal cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having renal cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having small cell lung cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having hepatocellular carcinoma. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having hypopharyngeal squamous cell carcinoma. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having neuroblastoma. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having breast cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having endometrial cancer. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having urothelial cell carcinoma. In one embodiment, the antibodies or ADCs of the invention are administered to a patient having acute myeloid leukemia (AML). In one embodiment, the antibodies or ADCs of the invention are administered to a patient having non-Hodgkin's lymphoma (NHL).
The term“B7-H3 expressing tumor,” as used herein, refers to a tumor which expresses B7-H3 protein. In one embodiment, B7-H3 expression in a tumor is determined using immunohistochemical staining of tumor cell membranes, where any immunohistochemical staining above background level in a tumor sample indicates that the tumor is a B7-H3 expressing tumor. Methods for detecting expression of B7-H3 in a tumor are known in the art, and include immunohistochemical assays. In contrast, a“B7-H3 negative tumor” is defined as a tumor having an absence of B7-H3 membrane staining above background in a tumor sample as determined by immunohistochemical techniques.
The terms“overexpress,”“overexpression,” or“overexpressed” interchangeably refer to a gene that is transcribed or translated at a detectably greater level, usually in a cancer cell, in comparison to a normal cell. Overexpression therefore refers to both overexpression of protein and RNA (due to increased transcription, post transcriptional processing, translation, post translational processing, altered stability, and altered protein degradation), as well as local overexpression due to altered protein traffic patterns (increased nuclear localization), and augmented functional activity, e.g., as in an increased enzyme hydrolysis of substrate. Thus, overexpression refers to either protein or RNA levels. Overexpression can also be by 50%, 60%, 70%, 80%, 90% or more in comparison to a normal cell or comparison cell. In certain embodiments, the anti-B7-H3 antibodies or ADCs of the invention are used to treat solid tumors likely to overexpress B7-H3 The term“gene amplification”, as used herein, refers to a cellular process characterized by the production of multiple copies of any particular piece of DNA. For example, a tumor cell may amplify, or copy, chromosomal segments as a result of cell signals and sometimes environmental events. The process of gene amplification leads to the production of additional copies of the gene. In one embodiment, the gene is B7-H3, i.e.,“B7-H3 amplification.” In one embodiment, the compositions and methods disclosed herein are used to treat a subject having B7-H3 amplified cancer.
The term“administering” as used herein is meant to refer to the delivery of a substance (e.g., an anti-B7-H3 antibody or ADC) to achieve a therapeutic objective (e.g., the treatment of a B7-H3- associated disorder). Modes of administration may be parenteral, enteral and topical. Parenteral administration is usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.
The term“combination therapy” or“combination” in the context of a therapeutic method (e.g., a treatment method),, as used herein, refers to the administration of two or more therapeutic substances, e.g., an anti-B7-H3 antibody or ADC and an additional therapeutic agent. The additional therapeutic agent may be administered concomitant with, prior to, or following the administration of the anti-B7-H3 antibody or ADC.
As used herein, the term“effective amount” or“therapeutically effective amount” refers to the amount of a drug, e.g., an antibody or ADC, which is sufficient to reduce or ameliorate the severity and/or duration of a disorder, e.g., cancer, or one or more symptoms thereof, prevent the advancement of a disorder, cause regression of a disorder, prevent the recurrence, development, onset or progression of one or more symptoms associated with a disorder, detect a disorder, or enhance or improve the prophylactic or therapeutic effect(s) of another therapy (e.g., prophylactic or therapeutic agent). The effective amount of an antibody or ADC may, for example, inhibit tumor growth (e.g., inhibit an increase in tumor volume), decrease tumor growth (e.g., decrease tumor volume), reduce the number of cancer cells, and/or relieve to some extent one or more of the symptoms associated with the cancer. The effective amount may, for example, improve disease free survival (DFS), improve overall survival (OS), or decrease likelihood of recurrence.
Various chemical substituents are defined below. In some instances, the number of carbon atoms in a substituent (e.g., alkyl, alkanyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, heteroaryl, and aryl) is indicated by the prefix“Cx-Cy” or“Cx-y” wherein x is the minimum and y is the maximum number of carbon atoms. Thus, for example,“C1-C6 alkyl” refers to an alkyl containing from 1 to 6 carbon atoms. Illustrating further,“C3-C8 cycloalkyl” means a saturated hydrocarbyl ring containing from 3 to 8 carbon ring atoms. If a substituent is described as being“substituted,” a hydrogen atom on a carbon or nitrogen is replaced with a non-hydrogen group. For example, a substituted alkyl substituent is an alkyl substituent in which at least one hydrogen atom on the alkyl is replaced with a non-hydrogen group. To illustrate, monofluoroalkyl is alkyl substituted with a fluoro radical, and difluoroalkyl is alkyl substituted with two fluoro radicals. It should be recognized that if there is more than one substitution on a substituent, each substitution may be identical or different (unless otherwise stated). If a substituent is described as being“optionally substituted”, the substituent may be either (1) not substituted or (2) substituted. Possible substituents include, but are not limited to, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, aryl, cycloalkyl, heterocyclyl, heteroaryl, halogen, C1-C6 haloalkyl, oxo, -CN, NO2, -ORxa, -OC(O)Rxz, -OC(O)N(Rxa)2, -SRxa, -S(O)2Rxa, -S(O)2N(Rxa)2, -C(O)Rxa, -C(O)ORxa, -C(O)N(Rxa)2, -C(O)N(Rxa)S(O)2Rxz, -N(Rxa)2, -N(Rxa)C(O)Rxz, -N(Rxa)S(O)2Rxz, -N(Rxa)C(O)O(Rxz), -N(Rxa)C(O)N(Rxa)2, -N(Rxa)S(O)2N(Rxa)2, -(C1-C6 alkylenyl)-CN, -(C1-C6 alkylenyl)-ORxa, -(C1-C6 alkylenyl)-OC(O)Rxz, -(C1-C6 alkylenyl)-OC(O)N(Rxa)2, -(C1-C6 alkylenyl)-SRxa, -(C1-C6
alkylenyl)-S(O)2Rxa, -(C1-C6 alkylenyl)-S(O)2N(Rxa)2, -(C1-C6 alkylenyl)-C(O)Rxa, -(C1-C6 alkylenyl)-C(O)ORxa, -(C1-C6 alkylenyl)-C(O)N(Rxa)2, -(C1-C6 alkylenyl)-C(O)N(Rxa)S(O)2Rxz, -(C1-C6 alkylenyl)-N(Rxa)2, -(C1-C6 alkylenyl)-N(Rxa)C(O)Rxz, -(C1-C6 alkylenyl)-N(Rxa)S(O)2Rxz, -(C1-C6 alkylenyl)-N(Rxa)C(O)O(Rxz), -(C1-C6 alkylenyl)-N(Rxa)C(O)N(Rxa)2, or -(C1-C6
alkylenyl)-N(Rxa)S(O)2N(Rxa)2; wherein Rxa, at each occurrence, is independently hydrogen, aryl, cycloalkyl, heterocyclyl, heteroaryl, C1-C6 alkyl, or C1-C6 haloalkyl; and Rxz, at each occurrence, is independently aryl, cycloalkyl, heterocyclyl, heteroaryl, C1-C6 alkyl or C1-C6 haloalkyl.
Various ADCs, synthons and Bcl-xL inhibitors comprising the ADCs and/or synthons are described in some embodiments herein by reference to structural formulae including substituents. It is to be understood that the various groups comprising substituents may be combined as valence and stability permit. Combinations of substituents and variables envisioned by this disclosure are only those that result in the formation of stable compounds. As used herein, the term“stable” refers to compounds that possess stability sufficient to allow manufacture and that maintain the integrity of the compound for a sufficient period of time to be useful for the purpose detailed herein.
As used herein, the following terms are intended to have the following meanings: The term“alkoxy” refers to a group of the formula–ORxa, where Rxa` is an alkyl group. Representative alkoxy groups include methoxy, ethoxy, propoxy, tert-butoxy and the like.
The term“alkoxyalkyl” refers to an alkyl group substituted with an alkoxy group and may be represented by the general formula–RbORxa where Rb is an alkylene group and Rxa is an alkyl group.
The term“alkyl” by itself or as part of another substituent refers to a saturated or unsaturated branched, straight-chain or cyclic monovalent hydrocarbon radical that is derived by the removal of one hydrogen atom from a single carbon atom of a parent alkane, alkene or alkyne.
Typical alkyl groups include, but are not limited to, methyl; ethyls such as ethanyl, ethenyl, ethynyl; propyls such as propan-1-yl, propan-2-yl, cyclopropan-1-yl, prop-1-en-1-yl, prop-1-en-2-yl, prop-2-en-1-yl, cycloprop-1-en-1-yl; cycloprop-2-en-1-yl, prop-1-yn-1-yl , prop-2-yn-1-yl, etc.; butyls such as butan-1-yl butan-2-yl 2-methyl-propan-1-yl 2-methyl-propan-2-yl cyclobutan-1-yl but-1-en-1-yl, but-1-en-2-yl, 2-methyl-prop-1-en-1-yl, but-2-en-1-yl , but-2-en-2-yl, buta-1,3-dien-1-yl, buta-1,3-dien-2-yl, cyclobut-1-en-1-yl, cyclobut-1-en-3-yl,
cyclobuta-1,3-dien-1-yl, but-1-yn-1-yl, but-1-yn-3-yl, but-3-yn-1-yl, etc.; and the like. Where specific levels of saturation are intended, the nomenclature“alkanyl,”“alkenyl” and/or“alkynyl” are used, as defined below. The term“lower alkyl” refers to alkyl groups with 1 to 6 carbons.
The term“alkanyl” by itself or as part of another substituent refers to a saturated branched, straight-chain or cyclic alkyl derived by the removal of one hydrogen atom from a single carbon atom of a parent alkane. Typical alkanyl groups include, but are not limited to, methyl; ethanyl; propanyls such as propan-1-yl, propan-2-yl (isopropyl), cyclopropan-1-yl, etc.; butanyls such as butan-1-yl, butan-2-yl (sec-butyl), 2-methyl-propan-1-yl (isobutyl), 2-methyl-propan-2-yl (t-butyl),
cyclobutan-1-yl, etc.; and the like.
The term“alkenyl” by itself or as part of another substituent refers to an unsaturated branched, straight-chain or cyclic alkyl having at least one carbon-carbon double bond derived by the removal of one hydrogen atom from a single carbon atom of a parent alkene. Typical alkenyl groups include, but are not limited to, ethenyl; propenyls such as prop-1-en-1-yl , prop-1-en-2-yl, prop-2-en-1-yl, prop-2-en-2-yl, cycloprop-1-en-1-yl; cycloprop-2-en-1-yl ; butenyls such as but-1-en-1-yl, but-1-en-2-yl, 2-methyl-prop-1-en-1-yl, but-2-en-1-yl, but-2-en-2-yl,
buta-1,3-dien-1-yl, buta-1,3-dien-2-yl, cyclobut-1-en-1-yl, cyclobut-1-en-3-yl,
cyclobuta-1,3-dien-1-yl, etc.; and the like.
The term“alkynyl” by itself or as part of another substituent refers to an unsaturated branched, straight-chain or cyclic alkyl having at least one carbon-carbon triple bond derived by the removal of one hydrogen atom from a single carbon atom of a parent alkyne. Typical alkynyl groups include, but are not limited to, ethynyl; propynyls such as prop-1-yn-1-yl , prop-2-yn-1-yl, etc.;
butynyls such as but-1-yn-1-yl, but-1-yn-3-yl, but-3-yn-1-yl , etc.; and the like.
The term“alkylamine” refers to a group of the formula -NHRxa and“dialkylamine” refers to a group of the formula–NRxaRxa, where each Rxa is, independently of the others, an alkyl group.
The term“alkylene” refers to an alkane, alkene or alkyne group having two terminal monovalent radical centers derived by the removal of one hydrogen atom from each of the two terminal carbon atoms. Typical alkylene groups include, but are not limited to, methylene; and saturated or unsaturated ethylene; propylene; butylene; and the like. The term“lower alkylene” refers to alkylene groups with 1 to 6 carbons.
The term“heteroalkylene” refers to a divalent alkylene having one or more—CH2— groups replaced with a thio, oxy, or—NRx3— where Rx3 is selected from hydrogen, lower alkyl and lower heteroalkyl. The heteroalkylene can be linear, branched, cyclic, bicyclic, or a combination thereof and can include up to 10 carbon atoms and up to 4 heteroatoms. The term“lower
heteroalkylene” refers to alkylene groups with 1 to 4 carbon atoms and 1 to 3 heteroatoms. The term“aryl” means an aromatic carbocyclyl containing from 6 to 14 carbon ring atoms. An aryl may be monocyclic or polycyclic (i.e., may contain more than one ring). In the case of polycyclic aromatic rings, only one ring the polycyclic system is required to be aromatic while the remaining ring(s) may be saturated, partially saturated or unsaturated. Examples of aryls include phenyl, naphthalenyl, indenyl, indanyl, and tetrahydronaphthyl.
The term“arylene” refers to an aryl group having two monovalent radical centers derived by the removal of one hydrogen atom from each of the two ring carbons. An exemplary arylene group is a phenylene.
An alkyl group may be substituted by a“carbonyl” which means that two hydrogen atoms from a single alkanylene carbon atom are removed and replaced with a double bond to an oxygen atom.
The prefix“halo” indicates that the substituent which includes the prefix is substituted with one or more independently selected halogen radicals. For example, haloalkyl means an alkyl substituent in which at least one hydrogen radical is replaced with a halogen radical. Typical halogen radicals include chloro, fluoro, bromo and iodo. Examples of haloalkyls include chloromethyl, 1- bromoethyl, fluoromethyl, difluoromethyl, trifluoromethyl, and 1,1,1-trifluoroethyl. It should be recognized that if a substituent is substituted by more than one halogen radical, those halogen radicals may be identical or different (unless otherwise stated).
The term“haloalkoxy” refers to a group of the formula–ORc, where Rc is a haloalkyl. The terms“heteroalkyl,”“heteroalkanyl,”“heteroalkenyl,”“heteroalkynyl,” and “heteroalkylene” refer to alkyl, alkanyl, alkenyl, alkynyl, and alkylene groups, respectively, in which one or more of the carbon atoms, e.g., 1, 2 or 3 carbon atoms, are each independently replaced with the same or different heteroatoms or heteroatomic groups. Typical heteroatoms and/or heteroatomic groups which can replace the carbon atoms include, but are not limited to, -O-, -S-, -S-O-, -NRc-, -PH, -S(O)-, -S(O)2-, -S(O)NRc-, -S(O)2NRc-, and the like, including combinations thereof, where each Rc is independently hydrogen or C1-C6 alkyl. The term“lower heteroalkyl” refers to between 1 and 4 carbon atoms and between 1 and 3 heteroatoms.
The terms“cycloalkyl” and“heterocyclyl” refer to cyclic versions of“alkyl” and “heteroalkyl” groups, respectively. For heterocyclyl groups, a heteroatom can occupy the position that is attached to the remainder of the molecule. A cycloalkyl or heterocyclyl ring may be a single- ring (monocyclic) or have two or more rings (bicyclic or polycyclic).
Monocyclic cycloalkyl and heterocyclyl groups will typically contains from 3 to 7 ring atoms, more typically from 3 to 6 ring atoms, and even more typically 5 to 6 ring atoms. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl; cyclobutyls such as cyclobutanyl and cyclobutenyl; cyclopentyls such as cyclopentanyl and cyclopentenyl; cyclohexyls such as
cyclohexanyl and cyclohexenyl; and the like. Examples of monocyclic heterocyclyls include, but are not limited to oxetane furanyl dihydrofuranyl tetrahydrofuranyl tetrahydropyranyl thiophenyl (thiofuranyl), dihydrothiophenyl, tetrahydrothiophenyl, pyrrolyl, pyrrolinyl, pyrrolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, triazolyl, tetrazolyl, oxazolyl, oxazolidinyl, isoxazolidinyl, isoxazolyl, thiazolyl, isothiazolyl, thiazolinyl, isothiazolinyl, thiazolidinyl, isothiazolidinyl, thiodiazolyl, oxadiazolyl (including 1,2,3-oxadiazolyl, 1,2,4- oxadiazolyl, 1,2,5-oxadiazolyl (furazanyl), or 1,3,4-oxadiazolyl), oxatriazolyl (including 1,2,3,4- oxatriazolyl or 1,2,3,5-oxatriazolyl), dioxazolyl (including 1,2,3-dioxazolyl, 1,2,4-dioxazolyl, 1,3,2- dioxazolyl, or 1,3,4-dioxazolyl), 1,4-dioxanyl, dioxothiomorpholinyl, oxathiazolyl, oxathiolyl, oxathiolanyl, pyranyl, dihydropyranyl, thiopyranyl, tetrahydrothiopyranyl, pyridinyl (azinyl), piperidinyl, diazinyl (including pyridazinyl (1,2-diazinyl), pyrimidinyl (1,3-diazinyl), or pyrazinyl (1,4-diazinyl)), piperazinyl, triazinyl (including 1,3,5-triazinyl, 1,2,4-triazinyl, and 1,2,3-triazinyl)), oxazinyl (including 1,2-oxazinyl, 1,3-oxazinyl, or 1,4-oxazinyl)), oxathiazinyl (including 1,2,3- oxathiazinyl, 1,2,4-oxathiazinyl, 1,2,5-oxathiazinyl, or 1,2,6-oxathiazinyl)), oxadiazinyl (including 1,2,3-oxadiazinyl, 1,2,4-oxadiazinyl, 1,4,2-oxadiazinyl, or 1,3,5-oxadiazinyl)), morpholinyl, azepinyl, oxepinyl, thiepinyl, diazepinyl, pyridonyl (including pyrid-2(1H)-onyl and pyrid-4(1H)-onyl), furan- 2(5H)-onyl, pyrimidonyl (including pyramid-2(1H)-onyl and pyramid-4(3H)-onyl), oxazol-2(3H)- onyl, 1H-imidazol-2(3H)-onyl, pyridazin-3(2H)-onyl, and pyrazin-2(1H)-onyl.
Polycyclic cycloalkyl and heterocyclyl groups contain more than one ring, and bicyclic cycloalkyl and heterocyclyl groups contain two rings. The rings may be in a bridged, fused or spiro orientation. Polycyclic cycloalkyl and heterocyclyl groups may include combinations of bridged, fused and/or spiro rings. In a spirocyclic cycloalkyl or heterocyclyl, one atom is common to two different rings. An example of a spirocycloalkyl is spiro[4.5]decane and an example of a
spiroheterocyclyls is a spiropyrazoline.
In a bridged cycloalkyl or heterocyclyl, the rings share at least two common non-adjacent atoms. Examples of bridged cycloalkyls include, but are not limited to, adamantyl and norbornanyl rings. Examples of bridged heterocyclyls include, but are not limited to, 2- oxatricyclo[3.3.1.13,7]decanyl.
In a fused-ring cycloalkyl or heterocyclyl, two or more rings are fused together, such that two rings share one common bond. Examples of fused-ring cycloalkyls include decalin, naphthylene, tetralin, and anthracene. Examples of fused-ring heterocyclyls containing two or three rings include imidazopyrazinyl (including imidazo[1,2-a]pyrazinyl), imidazopyridinyl (including imidazo[1,2- a]pyridinyl), imidazopyridazinyl (including imidazo[1,2-b]pyridazinyl), thiazolopyridinyl (including thiazolo[5,4-c]pyridinyl, thiazolo[5,4-b]pyridinyl, thiazolo[4,5-b]pyridinyl, and thiazolo[4,5- c]pyridinyl), indolizinyl, pyranopyrrolyl, 4H-quinolizinyl, purinyl, naphthyridinyl, pyridopyridinyl (including pyrido[3,4-b]-pyridinyl, pyrido[3,2-b]-pyridinyl, or pyrido[4,3-b]-pyridinyl), and pteridinyl. Other examples of fused-ring heterocyclyls include benzo-fused heterocyclyls, such as dihydrochromenyl, tetrahydroisoquinolinyl, indolyl, isoindolyl (isobenzazolyl, pseudoisoindolyl), indoleninyl (pseudoindolyl) isoindazolyl (benzpyrazolyl) benzazinyl (including quinolinyl (1- benzazinyl) or isoquinolinyl (2-benzazinyl)), phthalazinyl, quinoxalinyl, quinazolinyl, benzodiazinyl (including cinnolinyl (1,2-benzodiazinyl) or quinazolinyl (1,3-benzodiazinyl)), benzopyranyl (including chromanyl or isochromanyl), benzoxazinyl (including 1,3,2-benzoxazinyl, 1,4,2- benzoxazinyl, 2,3,1-benzoxazinyl, or 3,1,4-benzoxazinyl), benzo[d]thiazolyl, and benzisoxazinyl (including 1,2-benzisoxazinyl or 1,4-benzisoxazinyl).
The term“heteroaryl” refers to an aromatic heterocyclyl containing from 5 to 14 ring atoms. A heteroaryl may be a single ring or 2 or 3 fused rings. Examples of heteroaryls include 6- membered rings such as pyridyl, pyrazyl, pyrimidinyl, pyridazinyl, and 1,3,5-, 1,2,4- or 1,2,3- triazinyl; 5-membered ring substituents such as triazolyl, pyrrolyl, imidazyl, furanyl, thiophenyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, 1,2,3-, 1,2,4-, 1,2,5-, or 1,3,4-oxadiazolyl and isothiazolyl; 6/5-membered fused ring substituents such as imidazopyrazinyl (including imidazo[1,2- a]pyrazinyl)imidazopyridinyl (including imidazo[1,2-a]pyridinyl), imidazopyridazinyl (including imidazo[1,2-b]pyridazinyl), thiazolopyridinyl (including thiazolo[5,4-c]pyridinyl, thiazolo[5,4- b]pyridinyl, thiazolo[4,5-b]pyridinyl, and thiazolo[4,5-c]pyridinyl), benzo[d]thiazolyl,
benzothiofuranyl, benzisoxazolyl, benzoxazolyl, purinyl, and anthranilyl; and 6/6-membered fused rings such as benzopyranyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, and benzoxazinyl. Heteroaryls may also be heterocycles having aromatic (4N+2 pi electron) resonance contributors such as pyridonyl (including pyrid-2(1H)-onyl and pyrid-4(1H)-onyl), pyrimidonyl (including pyramid- 2(1H)-onyl and pyramid-4(3H)-onyl), pyridazin-3(2H)-onyl and pyrazin-2(1H)-onyl.
The term“sulfonate” as used herein means a salt or ester of a sulfonic acid.
The term“methyl sulfonate” as used herein means a methyl ester of a sulfonic acid group. The term“carboxylate” as used herein means a salt or ester of a carboxylic acid. The term“polyol”, as used herein, means a group containing more than two hydroxyl groups independently or as a portion of a monomer unit. Polyols include, but are not limited to, reduced C2-C6 carbohydrates, ethylene glycol, and glycerin.
The term“sugar” when used in context of“G1” includes O-glycoside, N-glycoside, S- glycoside and C-glycoside (C-glycoslyl) carbohydrate derivatives of the monosaccharide and disaccharide classes and may originate from naturally-occurring sources or may be synthetic in origin. For example“sugar” when used in context of“G1”includes derivatives such as but not limited to those derived from glucuronic acid, galacturonic acid, galactose, and glucose among others. Suitable sugar substitutions include but are not limited to hydroxyl, amine, carboxylic acid, sulfonic acid, phosphonic acid, esters, and ethers.
The term“NHS ester” means the N-hydroxysuccinimide ester derivative of a carboxylic acid.
The term“amine” includes primary, secondary and tertiary aliphatic amines, including cyclic versions. The term salt when used in context of“or salt thereof” include salts commonly used to form alkali metal salts and to form addition salts of free acids or free bases. In general, these salts typically may be prepared by conventional means by reacting, for example, the appropriate acid or base with a compound of the invention
Where a salt is intended to be administered to a patient (as opposed to, for example, being in use in an in vitro context), the salt preferably is pharmaceutically acceptable and/or physiologically compatible. The term "pharmaceutically acceptable" is used adjectivally in this patent application to mean that the modified noun is appropriate for use as a pharmaceutical product or as a part of a pharmaceutical product. The term“pharmaceutically acceptable salt” includes salts commonly used to form alkali metal salts and to form addition salts of free acids or free bases. In general, these salts typically may be prepared by conventional means by reacting, for example, the appropriate acid or base with a compound of the invention.
Various aspects of the invention are described in further detail in the following subsections. II. Anti-B7-H3 Antibodies
One aspect of the invention provides anti-B7-H3 antibodies, or antigen binding portions thereof. In one embodiment, the present invention provides chimeric anti-B7-H3 antibodies, or antigen binding portions thereof. In yet another embodiment, the present invention provides humanized anti-B7-H3 antibodies, or antigen binding portions thereof. In another aspect, the invention features antibody drug conjugates (ADCs) comprising an anti-B7-H3 antibody described herein and at least one drug(s), such as, but not limited to, a Bcl-xL inhibitor or an auristatin. The antibodies or ADCs of the invention have characteristics including, but not limited to, binding to wild- type human B7-H3 in vitro, binding to wild-type human B7-H3 on tumor cells expressing B7-H3, and decreasing or inhibiting xenograft tumor growth in a mouse model.
One aspect of the invention features an anti-human B7-H3 (anti-hB7-H3) Antibody Drug Conjugate (ADC) comprising an anti-hB7-H3 antibody conjugated to a drug via a linker, wherein the drug is a Bcl-xL inhibitor. Exemplary anti-B7-H3 antibodies (and sequences thereof) that can be used in the ADCs described herein.
The anti-B7-H3 antibodies described herein provide the ADCs of the invention with the ability to bind to B7-H3 such that the cytotoxic Bcl-xL drug attached to the antibody may be delivered to the B7-H3-expressing cell, particularly a B7-H3 expressing cancer cell.
While the term“antibody” is used throughout, it should be noted that antibody fragments (i.e., antigen-binding portions of an anti-B7-H3antibody) are also included in the invention and may be included in the embodiments (methods and compositions) described throughout. For example, an anti-B7-H3antibody fragment may be conjugated to the Bcl-xL inhibitors described herein. Thus, it is within the scope of the invention that in certain embodiments, antibody fragments of the anti-B7- H3antibodies described herein are conjugated to Bcl-xL inhibitors (including those described below in Section III.A) via linkers (including those described below in Section III.A). In certain embodiments, the anti-B7-H3 antibody binding portion is a Fab, a Fab’, a F(ab’)2, a Fv, a disulfide linked Fv, an scFv, a single domain antibody, or a diabody. II.A. Anti-B7-H3 Chimeric Antibodies
A chimeric antibody is a molecule in which different portions of the antibody are derived from different animal species, such as antibodies having a variable region derived from a murine monoclonal antibody and a human immunoglobulin constant region. Methods for producing chimeric antibodies are known in the art. See e.g., Morrison, Science 229:1202 (1985); Oi et al., BioTechniques 4:214 (1986); Gillies et al., (1989) J. Immunol. Methods 125:191-202; U.S. Pat. Nos.5,807,715; 4,816,567; and 4,816,397, which are incorporated herein by reference in their entireties. In addition, techniques developed for the production of "chimeric antibodies” (Morrison et al., 1984, Proc. Natl. Acad. Sci.81:851-855; Neuberger et al., 1984, Nature 312:604-608; Takeda et al., 1985, Nature 314:452-454, each of which are incorporated herein by reference in their entireties) by splicing genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity can be used.
As described in Example 3, eighteen anti-B7-H3 murine antibodies were identified having high specific binding activity against human and cynomolgus B7-H3. Chimeric antibodies, in the context of a human immunoglobulin constant region, were generated from these eighteen antibodies.
Thus, in one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence set forth in SEQ ID NOs: 1, 9, 16, 24, 32, 40, 48, 56, 64, 72, 80, 87, 95, 101, or 108; and/or a light chain variable region including an amino acid sequence set forth in SEQ ID NOs: 5, 13, 20, 28, 36, 44, 52, 60, 68, 76, 84, 91, 98, 105, or 112.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 1, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 5.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 2; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 3; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 4; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 6; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 8.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 9, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 13.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 11; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 14 (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 16, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 20.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 17; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 18; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 19; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 21; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 22; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 23.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 24, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 28.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 25; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 26; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 27; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 29; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 30; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 31.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 32, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 36.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 33; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 34; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 35; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 37; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 38; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 182.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 40, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 44.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 41; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 42; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 43; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 45; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 46; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 47.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 48, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 52.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 49; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 50; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 51; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 53; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 54; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 55.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 56, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 60.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 57; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 58; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 59; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 61; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 62; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 63.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 64, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 68.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 65; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 66; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 67; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 69; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 70; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 71.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 72, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 76.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 73; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 74; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 75; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 77; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 78; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 79.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 80, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 84.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 81; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 82; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 83; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 85; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 86.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 87 and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 91.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 88; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 89; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 90; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 92; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 93; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 94.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 95, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 98.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 49; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 96; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 97; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 99; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 93; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 100.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 101, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 105.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 102; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 103; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 104; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 106; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 46; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 107.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 108, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 112.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 109; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 110; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 111; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 113; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 114; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 115. II.B. Humanized Anti-B7-H3 Antibodies
The chimeric antibodies disclosed herein may be used in the production of humanized anti- B7-H3 antibodies. For example, following the generation and characterization of chimeric anti-B7- H3 antibodies chAb1-chAb18, antibodies chAb3, chAb13, and chAb18 were selected for
humanization. Specifically, six different humanized antibodies were created based on chAb3 (referred to herein as huAb3v1, huAb3v2, huAb3v3, huAb3v4, huAb3v5, and huAb3v6 (see
Examples 12 and 13), nine different humanized antibodies were created based on chAb13 (referred to herein as huAb13v1, huAb13v2, huAb13v3, huAb13v4, huAb13v5, huAb13v6, huAb13v7, huAb13v8, huAb13v9), and ten different humanized antibodies were created based on chAb18 (referred to herein as huAb18v1, huAb18v2, huAb18v3, huAb18v4, huAb18v5, huAb18v6, huAb18v7, huAb18v8, huAb18v9, and huAb18v10 (see Examples 9 and 10)). Tables 8, 12, 16, 18, and 19 provide the amino acid sequences of CDR, VH and VL regions of humanized chAb3, chAb13, and chAb18, respectively.
Generally, humanized antibodies are antibody molecules from non-human species antibody that binds the desired antigen having one or more complementarity determining regions (CDRs) from the non-human species and framework regions from a human immunoglobulin molecule. Known human Ig sequences are disclosed, e.g., www.ncbi.nlm.nih.gov/entrez- /query.fcgi;
www.atcc.org/phage/hdb.html; www.sciquest.com/; www.abcam.com/;
www.antibodyresource.com/onlinecomp.html;
www.public.iastate.edu/.about.pedro/research_tools.html; www.mgen.uni- heidelberg.de/SD/IT/IT.html; www.whfreeman.com/immunology/CH- 05/kuby05.htm;
www.library.thinkquest.org/12429/Immune/Antibody.html;
www.hhmi.org/grants/lectures/1996/vlab/; www.path.cam.ac.uk/.about.mrc7/m- ikeimages.html; www.antibodyresource.com/; mcb.harvard.edu/BioLinks/Immuno- logy.html.www.immunologylink.com/; pathbox.wustl.edu/.about.hcenter/index.- html;
www.biotech.ufl.edu/.about.hcl/; www.pebio.com/pa/340913/340913.html- ;
www.nal.usda.gov/awic/pubs/antibody/; www.m.ehime-u.acjp/.about.yasuhito- /Elisa.html;
www.biodesign.com/table.asp; www.icnet.uk/axp/facs/davies/lin- ks.html;
www.biotech.ufl.edu/.about.fccl/protocol.html; www.isac-net.org/sites_geo.html; aximtl.imt.uni- marburg.de/.about.rek/AEP- Start.html; baserv.uci.kun.nl/.about.jraats/linksl.html; www.recab.uni- hd.de/immuno.bme.nwu.edu/; www.mrc-cpe.cam.ac.uk/imt-doc/pu- blic/INTRO.html;
www ibt unam mx/vir/V mice html; imgt cnusc fr:8104/; www.biochem.ucl.ac.uk/.about.martin/abs/index.html; antibody.bath.ac.uk/;
abgen.cvm.tamu.edu/lab/wwwabgen.html; www.unizh.ch/.about.honegger/AHOsem- inar/Slide01.html; www.cryst.bbk.ac.uk/.about.ubcg07s/;
www.nimr.mrc.ac.uk/CC/ccaewg/ccaewg.htm; www.path.cam.ac.uk/.about.mrc7/h- umanisation/TAHHP.html; www.ibt.unam.mx/vir/structure/stat_aim.html;
www.biosci.missouri.edu/smithgp/index.html; www.cryst.bioc.cam.ac.uk/.abo- ut.fmolina/Web- pages/Pept/spottech.html; www.jerini.de/fr roducts.htm; www.patents.ibm.com/ibm.html.Kabat et al., Sequences of Proteins of Immunological Interest, U.S. Dept. Health (1983), each entirely incorporated herein by reference. Such imported sequences can be used to reduce immunogenicity or reduce, enhance or modify binding, affinity, on-rate, off-rate, avidity, specificity, half-life, or any other suitable characteristic, as known in the art.
Framework residues in the human framework regions may be substituted with the corresponding residue from the CDR donor antibody to alter, preferably improve, antigen binding. These framework substitutions are identified by methods well known in the art, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions. (See, e.g., Queen et al., U.S. Pat. No.5,585,089; Riechmann et al., Nature 332:323 (1988), which are incorporated herein by reference in their entireties.) Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art. Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen. In this way, FR residues can be selected and combined from the consensus and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen(s), is achieved. In general, the CDR residues are directly and most substantially involved in influencing antigen binding. Antibodies can be humanized using a variety of techniques known in the art, such as but not limited to those described in Jones et al., Nature 321:522 (1986); Verhoeyen et al., Science 239:1534 (1988)), Sims et al., J. Immunol.151: 2296 (1993); Chothia and Lesk, J. Mol. Biol.196:901 (1987), Carter et al., Proc. Natl. Acad. Sci. U.S.A.89:4285 (1992); Presta et al., J. Immunol.151:2623 (1993), Padlan, Molecular Immunology 28(4/5):489-498 (1991); Studnicka et al., Protein Engineering 7(6):805-814 (1994); Roguska. et al. , PNAS 91:969-973 (1994); PCT publication WO 91/09967, PCT/:
US98/16280, US96/18978, US91/09630, US91/05939, US94/01234, GB89/01334, GB91/01134, GB92/01755; WO90/14443, WO90/14424, WO90/14430, EP 229246, EP 592,106; EP 519,596, EP 239,400, U.S. Pat. Nos.5,565,332, 5,723,323, 5,976,862, 5,824,514, 5,817,483, 5814476, 5763192, 5723323, 5,766886, 5,714,352, 6,204,023, 6,180,370, 5,693,762, 5,530,101, 5,585,089, 5,225,539; 4816567 each entirely incorporated herein by reference included references cited therein Humanized anti-B7-H3 antibodies derived from chAb3
Six humanized antibodies based on chAb3 were created. The sequences of each are as follows:
A) huAb3v1 (VH amino acid sequence set forth in SEQ ID NO: 125 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 11, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 128 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 14, 7, and 15, respectively);
B) huAb3v2 (VH amino acid sequence set forth in SEQ ID NO: 127 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 11, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 128 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 14, 7, and 15, respectively);
C) huAb3v3 (VH amino acid sequence set forth in SEQ ID NO: 126 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 11, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 129 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 14, 7, and 15, respectively);
D) huAb3v4 (VH amino acid sequence set forth in SEQ ID NO: 125 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 11, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 130 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 14, 7, and 15, respectively);
E) huAb3v5 (VH amino acid sequence set forth in SEQ ID NO: 127 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 11, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 130 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 14, 7, and 15, respectively); and
F) huAb3v6 (VH amino acid sequence set forth in SEQ ID NO: 126 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 11, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 130 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 14, 7, and 15, respectively).
Of the six humanized versions of chAb3, huAb3v2 was selected for further modified in order to remove potential deamidation or isomerization sites in the light chain CDR1 or in the heavy chain CDR2. Nine variants of the humanized antibody huAb3v2 were generated, and are referred to herein as huAb3v2.1, huAb3v2.2, huAb3v2.3, huAb3v2.4, huAb3v2.5, huAb3v2.6, huAb3v2.7, huAb3v2.8, and huAb3v2.9 (CDR and variable domain sequences are provided in Table 13). The nine variants of the huAb3v2 antibody include the following:
A) huAb3v2.1 (VH amino acid sequence set forth in SEQ ID NO: 131 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 132, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 133 and VL CDR1 CDR2 and CDR3 amino acid sequences set forth in SEQ ID NOs: 134, 7, and 15, respectively);
B) huAb3v2.2 (VH amino acid sequence set forth in SEQ ID NO: 131 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 132, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 135 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 136, 7, and 15, respectively);
C) huAb3v2.3 (VH amino acid sequence set forth in SEQ ID NO: 131 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 132, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 137 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 138, 7, and 15, respectively);
D) huAb3v2.4 (VH amino acid sequence set forth in SEQ ID NO: 139 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 140, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 133 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 134, 7, and 15, respectively);
E) huAb3v2.5 (VH amino acid sequence set forth in SEQ ID NO: 139 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 140, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 135 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 136, 7, and 15, respectively);
F) huAb3v2.6 (VH amino acid sequence set forth in SEQ ID NO: 139 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 140, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 137 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 138, 7, and 15, respectively);
G) huAb3v2.7 (VH amino acid sequence set forth in SEQ ID NO: 141 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 142, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 133 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 134, 7, and 15, respectively);
H) huAb3v2.8 (VH amino acid sequence set forth in SEQ ID NO: 141 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 142, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 135 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 136, 7, and 15, respectively); and
I) huAb3v2.9 (VH amino acid sequence set forth in SEQ ID NO: 141 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 10, 142, and 12, respectively; and VL amino acid sequence set forth in SEQ ID NO: 137 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 138, 7, and 15, respectively).
Thus, in one aspect, the present invention provides antibodies comprising variable and/or CDR sequences from a humanized antibody derived from chAb3. In one embodiment, the invention features anti-B7-H3 antibodies which are derived from Ab3 have improved characteristics, e.g., improved binding affinity to isolated B7-H3 protein and improved binding to B7-H3 expressing cells as described in the Examples below. Collectively these novel antibodies are referred to herein as “Ab3 variant antibodies.” Generally, the Ab3 variant antibodies retain the same epitope specificity as Ab3. In various embodiments, anti-B7-H3 antibodies, or antigen binding fragments thereof, of the invention are capable of modulating a biological function of B7-H3.
In one aspect, the present invention provides a humanized antibody, or antigen binding portion thereof, having a heavy chain variable region including an amino acid sequence set forth in SEQ ID NOs: 125, 126, 127, 131, 139, or 141; and/or a light chain variable region including an amino acid sequence set forth in SEQ ID NOs: 128, 129, 130, 133, 135, or 137.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen binding portion thereof, of the invention comprises a heavy chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 10; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 11, 132, 140, or 142; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 14, 134, 136, or 138; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 7; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 15.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 125, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 128.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 127, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 128.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 126, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 129.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 125, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 130.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 127, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 130.
In one aspect the present invention is directed to an anti-B7-H3 antibody or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 126, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 130.
In another aspect, the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 11; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 14; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 131, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 133.
In another aspect, the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 132; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 134; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 131, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 135.
In another aspect, the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 132; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 136; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 131, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 137.
In another aspect the present invention is directed to a humanized anti-B7-H3 antibody or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 132; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 138; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 139, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 133.
In another aspect, the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 140; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 134; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 139, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 135.
In another aspect, the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 140; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 136; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain comprising the amino acid sequence of SEQ ID NO: 170 and a light chain comprising the amino acid sequence of SEQ ID NO: 171. In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 139, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 137.
In another aspect, the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 140; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 138; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen- binding portion thereof, having a heavy chain comprising the amino acid sequence of SEQ ID NO: 172 and a light chain comprising the amino acid sequence of SEQ ID NO: 173.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 141, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 133
In another aspect, the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 142; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 134; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 141, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 135.
In another aspect, the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 142; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 136; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 141, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 137.
In another aspect, the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 142; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 138; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15. Humanized anti-B7-H3 antibodies derived from chAb13
The nine different humanized antibodies created based on chAb13 include the following: A) huAb13v1 (VH amino acid sequence set forth in SEQ ID NO: 147 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 33, 34, and 35, respectively; and VL amino acid sequence set forth in SEQ ID NO: 144 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 37, 38, and 39, respectively);
B) huAb13v2 (VH amino acid sequence set forth in SEQ ID NO: 146 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 33, 34, and 35, respectively; and VL amino acid sequence set forth in SEQ ID NO: 143 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 37, 38, and 39, respectively);
C) huAb13v3 (VH amino acid sequence set forth in SEQ ID NO: 146 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 33, 34, and 35, respectively; and VL amino acid sequence set forth in SEQ ID NO: 144 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 37, 38, and 39, respectively);
D) huAb13v4 (VH amino acid sequence set forth in SEQ ID NO: 146 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 33, 34, and 35, respectively; and VL amino acid sequence set forth in SEQ ID NO: 145 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 37, 38, and 39, respectively);
E) huAb13v5 (VH amino acid sequence set forth in SEQ ID NO: 147 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 33, 34, and 35, respectively; and VL amino acid sequence set forth in SEQ ID NO: 143 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 37, 38, and 39, respectively);
F) huAb13v6 (VH amino acid sequence set forth in SEQ ID NO: 147 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 33, 34, and 35, respectively; and VL amino acid sequence set forth in SEQ ID NO: 145 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 37, 38, and 39, respectively);
G) huAb13v7 (VH amino acid sequence set forth in SEQ ID NO: 148 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 33, 34, and 35, respectively; and VL amino acid sequence set forth in SEQ ID NO: 143 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 37, 38, and 39, respectively);
H) huAb13v8 (VH amino acid sequence set forth in SEQ ID NO: 148 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 33, 34, and 35, respectively; and VL amino acid sequence set forth in SEQ ID NO: 144 and VL CDR1 CDR2 and CDR3 amino acid sequences set forth in SEQ ID NOs: 37, 38, and 39, respectively);
I) huAb13v9 (VH amino acid sequence set forth in SEQ ID NO: 148 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 33, 34, and 35, respectively; and VL amino acid sequence set forth in SEQ ID NO: 145 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 37, 38, and 39, respectively).
Thus, in one aspect the present invention provides antibodies comprising variable and/or CDR sequences from a humanized antibody derived from chAb13. In one embodiment, the invention features anti-B7-H3 antibodies which are derived from chAb13 have improved characteristics, e.g., improved binding affinity to isolated B7-H3 protein and improved binding to B7-H3 expressing cells, as described in the Examples below. Collectively these novel antibodies are referred to herein as “Ab13 variant antibodies.” Generally, the Ab13 variant antibodies retain the same epitope specificity as Ab13. In various embodiments, anti-B7-H3 antibodies, or antigen binding fragments thereof, of the invention are capable of modulating a biological function of B7-H3.
In one aspect, the present invention provides a humanized antibody, or antigen binding portion thereof, having a heavy chain variable region including an amino acid sequence set forth in SEQ ID NOs: 146, 147, or 148; and/or a light chain variable region including an amino acid sequence set forth in SEQ ID NOs: 143, 144, or 145.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen binding portion thereof, of the invention comprises a heavy chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 33; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 34; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 35; and a light chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 37; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 38; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 39.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 147, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 144. In one embodiment, the invention provides an anti-B7H3 antibody comprising the CDR sequences set forth in the variable regions of huAb13v1 (SEQ ID NOs.144 and 147).
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen binding portion thereof, having a heavy chain comprising the amino acid sequence of SEQ ID NO: 168 and a light chain comprising the amino acid sequence of SEQ ID NO: 169.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 146, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 143 In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 146, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 144.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 146, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 145.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 147, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 143.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 147, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 145.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 148, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 143.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 148, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 144.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 148, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 145. Humanized anti-B7-H3 antibodies derived from chAb18
The ten different humanized antibodies created based on chAb18 include the following: A) huAb18v1 (VH amino acid sequence set forth in SEQ ID NO: 116 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25, 26, and 27, respectively; and VL amino acid sequence set forth in SEQ ID NO: 120 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively);
B) huAb18v2 (VH amino acid sequence set forth in SEQ ID NO: 118 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25 119 and 27 respectively; and VL amino acid sequence set forth in SEQ ID NO: 120 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively);
C) huAb18v3 (VH amino acid sequence set forth in SEQ ID NO: 117 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25, 26, and 27, respectively; and VL amino acid sequence set forth in SEQ ID NO: 121 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively);
D) huAb18v4 (VH amino acid sequence set forth in SEQ ID NO: 118 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25, 119, and 27, respectively; and VL amino acid sequence set forth in SEQ ID NO: 121 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively);
E) huAb18v5 (VH amino acid sequence set forth in SEQ ID NO: 116 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25, 26, and 27, respectively; and VL amino acid sequence set forth in SEQ ID NO: 123 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively);
F) huAb18v6 (VH amino acid sequence set forth in SEQ ID NO: 118 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25, 119, and 27, respectively; and VL amino acid sequence set forth in SEQ ID NO: 123 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively);
G) huAb18v7 (VH amino acid sequence set forth in SEQ ID NO: 118 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25, 119, and 27, respectively; and VL amino acid sequence set forth in SEQ ID NO: 124 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively);
H) huAb18v8 (VH amino acid sequence set forth in SEQ ID NO: 117 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25, 26, and 27, respectively; and VL amino acid sequence set forth in SEQ ID NO: 122 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively);
I) huAb18v9 (VH amino acid sequence set forth in SEQ ID NO: 117 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25, 26, and 27, respectively; and VL amino acid sequence set forth in SEQ ID NO: 124 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively); and
J) huAb18v10 (VH amino acid sequence set forth in SEQ ID NO: 118 and VH CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 25, 119, and 27, respectively; and VL amino acid sequence set forth in SEQ ID NO: 122 and VL CDR1, CDR2, and CDR3 amino acid sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively).
Thus, in one aspect the present invention provides antibodies comprising variable and/or CDR sequences from a humanized antibody derived from chAb18. In one embodiment, the invention features anti-B7-H3 antibodies which are derived from Ab18 have improved characteristics e g improved binding affinity to isolated B7-H3 protein and improved binding to B7-H3 expressing cells, as described in the Examples below. Collectively these novel antibodies are referred to herein as “Ab18 variant antibodies.” Generally, the Ab18 variant antibodies retain the same epitope specificity as Ab18. In various embodiments, anti-B7-H3 antibodies, or antigen binding fragments thereof, of the invention are capable of modulating a biological function of B7-H3.
In one aspect, the present invention provides a humanized antibody, or antigen binding portion thereof, having a heavy chain variable region including an amino acid sequence set forth in SEQ ID NOs: 116, 117, or 118; and/or a light chain variable region including an amino acid sequence set forth in SEQ ID NOs: 120, 121, 122, 123 or 124.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen binding portion thereof, of the invention comprises a heavy chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 25; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 26 or 119; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 27; and a light chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 29; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 30; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 31.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 116, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 120.
In another aspect, the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 25; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 26; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 27; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 29; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 30; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 31.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 118, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 120.
In another aspect, the present invention is directed to a humanized anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 25; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 119; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 27; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 29; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 30; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 31.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 117, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 121.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 118, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 121.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 116, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 123.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 118, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 123.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 118, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 124.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 117, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 122.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 117, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 124.
In one aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen-binding portion thereof, having a heavy chain variable region including an amino acid sequence as set forth in SEQ ID NO: 118, and a light chain variable region including an amino acid sequence set forth in SEQ ID NO: 122.
In one aspect, the present invention provides a humanized antibody, or antigen binding portion thereof, having a heavy chain variable region including an amino acid sequence set forth in SEQ ID NOs: 116 117 118 146 147 148 125 126 127 131 139 or 141; and/or a light chain variable region including an amino acid sequence set forth in SEQ ID NOs: 120, 121, 122, 123, 124, 143, 144, 145, 128, 129, 130, 133, 135, or 137.
In another aspect, the present invention is directed to an anti-B7-H3 antibody, or antigen binding portion thereof, of the invention comprises a heavy chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 10, 25, or 33; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 11, 26, 34, 119, 132, 140, or 142; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 12, 27, or 35; and a light chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 14, 29, 37, 134, 136, or 138; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 7, 30, or 38; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 15, 31 or 39.
In another aspect, the invention provides an anti-B7-H3 antibody, or antigen binding fragment thereof, that specifically competes with an anti-B7-H3 antibody, or fragment thereof, described herein, wherein said competition can be detected in a competitive binding assay using said antibody, the human B7-H3 polypeptide, and the anti-B7-H3 antibody or fragment thereof. In particular embodiments, the competing antibody, or antigen binding portion thereof, is an antibody, or antigen binding portion thereof, that competes with huAb3v2.5, huAb3v2.6, or huAb13v1.
In one embodiment, the anti-B7-H3 antibodies, or antigen binding portions thereof, of the invention bind to the extracellular domain of human B7-H3 (SEQ ID NO: 152) with a dissociation constant (KD) of about 1 x 10-6 M or less, as determined by surface plasmon resonance. Alternatively, the antibodies, or antigen binding portions thereof, bind to human B7-H3 with a KD of between about 1 x 10-6 M and about 1 x 10-11 M, as determined by surface plasmon resonance. In a further alternative, antibodies, or antigen binding portions thereof, bind to human B7-H3 with a KD of between about 1 x 10-6 M and about 1 x 10-7 M, as determined by surface plasmon resonance.
Alternatively, antibodies, or antigen binding portions thereof, of the invention binds to human B7-H3 with a KD of between about 1 x 10-6 M and about 5 x 10-11 M, about 1 x 10-6 M and about 5 x 10-10 M; a KD of between about 1 x 10-6 M and about 1 x 10-9 M; a KD of between about 1 x 10-6 M and about 5 x 10-9 M; a KD of between about 1 x 10-6 M and about 1 x 10-8 M; a KD of between about 1 x 10-6 M and about 5 x 10-8 M; a KD of between about 8.4 x 10-7 M and about 3.4 x 10-11 M; a KD of between about 5.9 x 10-7 M; and about 2.2 x 10-7 M, as determined by surface plasmon resonance.
In one embodiment, the antibodies, or antigen binding portions thereof, of the invention bind to human B7-H3 (SEQ ID NO: 149) with a KD of about 1 x 10-6 M or less, as determined by surface plasmon resonance. Alternatively, the antibodies, or antigen binding portions thereof, of the invention bind to human B7-H3 (SEQ ID NO: 149) with a KD of between about 8.2 x 10-9 M and about 6.3 x 10- 10 M; a KD of between about 8.2 x 10-9 M and about 2.0 x 10-9 M; a KD of between about 2.3 x 10-9 M and about 1.5 x 10-10 M, as determined by surface plasmon resonance. Anti-B7-H3 antibodies provided herein may comprise a heavy chain variable region comprising CDR1, CDR2 and CDR3 sequences and a light chain variable region comprising CDR1, CDR2 and CDR3 sequences, wherein one or more of these CDR sequences comprise specified amino acid sequences based on the antibodies described herein (e.g., huAb13v1 or huAb3v2.5), or conservative modifications thereof, and wherein the antibodies retain the desired functional properties of the anti-B7-H3antibodies described herein. Accordingly, the anti-B7-H3 antibody, or antigen binding portion thereof, may comprise a heavy chain variable region comprising CDR1, CDR2, and CDR3 sequences and a light chain variable region comprising CDR1, CDR2, and CDR3 sequences, wherein: (a) the heavy chain variable region CDR3 sequence comprises SEQ ID NO: 12 or 35, and conservative modifications thereof, e.g., 1, 2, 3, 4, 5, 1-2, 1-3, 1-4 or 1-5 conservative amino acid substitutions; (b) the light chain variable region CDR3 sequence comprises SEQ ID NO: 15 or 39, and conservative modifications thereof, e.g., 1, 2, 3, 4, 5, 1-2, 1-3, 1-4 or 1-5 conservative amino acid substitutions; (c) the antibody specifically binds to B7-H3, and (d) the antibody exhibits 1, 2, 3, 4, 5, 6, or all of the following functional properties described herein, e.g., binding to soluble human B7-H3. In a one embodiment, the heavy chain variable region CDR2 sequence comprises SEQ ID NO: 140 or 34, and conservative modifications thereof, e.g., 1, 2, 3, 4, 5, 1-2, 1-3, 1-4 or 1-5 conservative amino acid substitutions; and the light chain variable region CDR2 sequence comprises SEQ ID NO: 7 or 38, and conservative modifications thereof, e.g., 1, 2, 3, 4, 5, 1-2, 1-3, 1-4 or 1-5 conservative amino acid substitutions. In another preferred embodiment, the heavy chain variable region CDR1 sequence comprises SEQ ID NO: 10 or 33, and conservative modifications thereof, e.g., 1, 2, 3, 4, 5, 1-2, 1-3, 1-4 or 1-5 conservative amino acid substitutions; and the light chain variable region CDR1 sequence comprises SEQ ID NO: 136, 138, or 37, and conservative modifications thereof, e.g., 1, 2, 3, 4, 5, 1-2, 1-3, 1-4 or 1-5 conservative amino acid substitutions.
Conservative amino acid substitutions may also be made in portions of the antibodies other than, or in addition to, the CDRs. For example, conservative amino acid modifications may be made in a framework region or in the Fc region. A variable region or a heavy or light chain may comprise 1, 2, 3, 4, 5, 1-2, 1-3, 1-4, 1-5, 1-10, 1-15, 1-20, 1-25, or 1-50 conservative amino acid substitutions relative to the anti-B7-H3 antibody sequences provided herein. In certain embodiments, the anti-B7- H3 antibody comprises a combination of conservative and non-conservative amino acid modification.
To generate and to select CDRs having preferred B7-H3 binding and/or neutralizing activity with respect to hB7-H3, standard methods known in the art for generating antibodies, or antigen binding portions thereof, and assessing the B7-H3 binding and/or neutralizing characteristics of those antibodies, or antigen binding portions thereof, may be used, including but not limited to those specifically described herein.
The foregoing establish a novel family of B7-H3 binding proteins, isolated in accordance with this invention, and including antigen binding polypeptides that comprise the CDR sequences listed in the Sequence Table provided herein To generate and to select CDRs having preferred B7-H3 binding and/or neutralizing activity with respect to hB7-H3, standardmethods known in the art for generating antibodies, or antigen binding portions thereof, and assessing the B7-H3 binding and/or neutralizing characteristics of those antibodies, or antigen binding portions thereof, may be used, including but not limited to those specifically described herein.
In certain embodiments, the antibody comprises a heavy chain constant region, such as an IgG1, IgG2, IgG3, IgG4, IgA, IgE, IgM, or IgD constant region. In certain embodiments, the anti-B7- H3 antibody, or antigen binding portion thereof, comprises a heavy chain immunoglobulin constant domain selected from the group consisting of a human IgG constant domain, a human IgM constant domain, a human IgE constant domain, and a human IgA constant domain. In further embodiments, the antibody, or antigen binding portion thereof, has an IgG1 heavy chain constant region, an IgG2 heavy chain constant region, an IgG3 constant region, or an IgG4 heavy chain constant region.
Preferably, the heavy chain constant region is an IgG1 heavy chain constant region or an IgG4 heavy chain constant region. Furthermore, the antibody can comprise a light chain constant region, either a kappa light chain constant region or a lambda light chain constant region. Preferably, the antibody comprises a kappa light chain constant region. Alternatively, the antibody portion can be, for example, a Fab fragment or a single chain Fv fragment.
In certain embodiments, the anti-B7-H3 antibody binding portion is a Fab, a Fab’, a F(ab’)2, a Fv, a disulfide linked Fv, an scFv, a single domain antibody, or a diabody.
In certain embodiments, the anti-B7-H3 antibody, or antigen binding portion thereof, is a multispecific antibody, e.g. a bispecific antibody.
Replacements of amino acid residues in the Fc portion to alter antibody effector function have been described (Winter, et al. US Patent Nos.5,648,260 and 5,624,821, incorporated by reference herein). The Fc portion of an antibody mediates several important effector functions e.g. cytokine induction, ADCC, phagocytosis, complement dependent cytotoxicity (CDC) and half-life/ clearance rate of antibody and antigen-antibody complexes. In some cases these effector functions are desirable for therapeutic antibody but in other cases might be unnecessary or even deleterious, depending on the therapeutic objectives. Certain human IgG isotypes, particularly IgG1 and IgG3, mediate ADCC and CDC via binding to FcγRs and complement C1q, respectively. Neonatal Fc receptors (FcRn) are the critical components determining the circulating half-life of antibodies. In still another embodiment at least one amino acid residue is replaced in the constant region of the antibody, for example the Fc region of the antibody, such that effector functions of the antibody are altered.
One embodiment of the invention includes a recombinant chimeric antigen receptor (CAR) comprising the binding regions of the antibodies described herein, e.g., the heavy and/or light chain CDRs of huAb13v1. A recombinant CAR, as described herein, may be used to redirect T cell specificity to an antigen in a human leukocyte antigen (HLA)-independent fashion. Thus, CARs of the invention may be used in immunotherapy to help engineer a human subject’s own immune cells to recognize and attack the subject’s tumor (see, e.g., U.S. Pat. Nos.6,410,319; 8,389,282; 8,822,647; 8,906,682; 8,911,993; 8,916,381; 8,975,071; and U.S. Patent Appln. Publ. No. US20140322275, each of which is incorporated by reference herein with respect to CAR technology). This type of immunotherapy is called adoptive cell transfer (ACT), and may be used to treat cancer in a subject in need thereof.
An anti-B7-H3 CAR of the invention preferably contains a extracellular antigen-binding domain specific for B7-H3, a transmembrane domain which is used to anchor the CAR into a T cell, and one or more intracellular signaling domains. In one embodiment of the invention, the CAR includes a transmembrane domain that comprises a transmembrane domain of a protein selected from the group consisting of the alpha, beta or zeta chain of the T-cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137 and CD154. In one embodiment of the invention, the CAR comprises a costimulatory domain, e.g., a costimulatory domain comprising a functional signaling domain of a protein selected from the group consisting of OX40, CD2, CD27, CD28, CD5, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), and 4-1BB (CD137). In certain embodiments of the invention, the CAR comprises an scFv comprising the CDR or variable regions described herein e.g., CDRs or variable regions from the huAb13v1 antibody, a transmembrane domain, a co-stimulatory domain (e.g., a functional signaling domain from CD28 or 4-1BB), and a signaling domain comprising a functional signaling domain from CD3 (e.g., CD3-zeta).
In certain embodiments, the invention incudes a T cell comprising a CAR (also referred to as a CAR T cell) comprising antigen binding regions, e.g. CDRs, of the antibodies described herein or an scFv described herein.
In certain embodiments of the invention, the CAR comprises a heavy chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 10, 25, or 33; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 11, 26, 34, 119, 132, 140, or 142; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 12, 27, or 35; and a light chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 14, 29, 37, 134, 136, or 138; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 7, 30, or 38; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 15, 31 or 39.
In certain embodiments of the invention, the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 11; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 14; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
In certain embodiments of the invention, the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 132; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 134; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
In certain embodiments of the invention, the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 132; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 136; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
In certain embodiments of the invention, the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 132; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 138; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
In certain embodiments of the invention, the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 140; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 134; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
In certain embodiments of the invention, the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 140; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 136; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15 In certain embodiments of the invention, the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 140; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 138; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
In certain embodiments of the invention, the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 142; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 134; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
In certain embodiments of the invention, the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 142; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 136; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15.
In certain embodiments of the invention, the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 10; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 142; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 12; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 138; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 7; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 15. In certain embodiments of the invention, the CAR comprises a heavy chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 33; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 34; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 35; and a light chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 37; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 38; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 39.
In certain embodiments of the invention, the CAR comprises a heavy chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 25; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 26 or 119; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 27; and a light chain variable region comprising a CDR1 domain comprising an amino acid sequence as set forth in SEQ ID NO: 29; a CDR2 domain comprising an amino acid sequence as set forth in SEQ ID NO: 30; and a CDR3 domain comprising an amino acid sequence as set forth in SEQ ID NO: 31.
In certain embodiments of the invention, the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 25; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 26; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 27; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 29; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 30; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 31.
In certain embodiments of the invention, the CAR comprises a heavy chain variable domain region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 25; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 119; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 27; and a light chain variable region including (a) a CDR1 having an amino acid sequence as set forth in SEQ ID NO: 29; (b) a CDR2 having an amino acid sequence as set forth in SEQ ID NO: 30; and (c) a CDR3 having an amino acid sequence as set forth in SEQ ID NO: 31.
One embodiment of the invention includes a labeled anti-B7-H3 antibody, or antibody portion thereof, where the antibody is derivatized or linked to one or more functional molecule(s) (e.g., another peptide or protein). For example, a labeled antibody can be derived by functionally linking an antibody or antibody portion of the invention (by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as another antibody (e.g., a bispecific antibody or a diabody), a detectable agent, a pharmaceutical agent, a protein or peptide that can mediate the association of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag), and/or a cytotoxic or therapeutic agent selected from the group consisting of a mitotic inhibitor, an antitumor antibiotic, an immunomodulating agent, a vector for gene therapy, an alkylating agent, an antiangiogenic agent, an antimetabolite, a boron- containing agent, a chemoprotective agent, a hormone, an antihormone agent, a corticosteroid, a photoactive therapeutic agent, an oligonucleotide, a radionuclide agent, a topoisomerase inhibitor, a kinase inhibitor, a radiosensitizer, and a combination thereof.
Useful detectable agents with which an antibody or antibody portion thereof, may be derivatized include fluorescent compounds. Exemplary fluorescent detectable agents include fluorescein, fluorescein isothiocyanate, rhodamine, 5-dimethylamine-1-napthalenesulfonyl chloride, phycoerythrin and the like. An antibody may also be derivatized with detectable enzymes, such as alkaline phosphatase, horseradish peroxidase, glucose oxidase and the like. When an antibody is derivatized with a detectable enzyme it is detected by adding additional reagents that the enzyme uses to produce a detectable reaction product. For example, when the detectable agent horseradish peroxidase is present the addition of hydrogen peroxide and diaminobenzidine leads to a colored reaction product, which is detectable. An antibody may also be derivatized with biotin, and detected through indirect measurement of avidin or streptavidin binding.
In one embodiment, the antibody of the invention is conjugated to an imaging agent.
Examples of imaging agents that may be used in the compositions and methods described herein include, but are not limited to, a radiolabel (e.g., indium), an enzyme, a fluorescent label, a luminescent label, a bioluminescent label, a magnetic label, and biotin.
In one embodiment, the antibodies or ADCs are linked to a radiolabel, such as, but not limited to, indium (111In). 111Indium may be used to label the antibodies and ADCs described herein for use in identifying B7-H3 positive tumors. In a certain embodiment, anti-B7-H3 antibodies (or ADCs) described herein are labeled with 111I via a bifunctional chelator which is a bifunctional cyclohexyl diethylenetriaminepentaacetic acid (DTPA) chelate (see US Patent Nos.5,124,471; 5,434,287; and 5,286,850, each of which is incorporated herein by reference).
Another embodiment of the invention provides a glycosylated binding protein wherein the anti-B7-H3 antibody or antigen binding portion thereof comprises one or more carbohydrate residues. Nascent in vivo protein production may undergo further processing, known as post-translational modification. In particular, sugar (glycosyl) residues may be added enzymatically, a process known as glycosylation. The resulting proteins bearing covalently linked oligosaccharide side chains are known as glycosylated proteins or glycoproteins. Antibodies are glycoproteins with one or more carbohydrate residues in the Fc domain, as well as the variable domain. Carbohydrate residues in the Fc domain have important effect on the effector function of the Fc domain, with minimal effect on antigen binding or half-life of the antibody (R. Jefferis, Biotechnol. Prog.21 (2005), pp.11–16). In contrast, glycosylation of the variable domain may have an effect on the antigen binding activity of the antibody. Glycosylation in the variable domain may have a negative effect on antibody binding affinity, likely due to steric hindrance (Co, M.S., et al., Mol. Immunol. (1993) 30:1361- 1367), or result in increased affinity for the antigen (Wallick, S.C., et al., Exp. Med. (1988) 168:1099-1109; Wright, A., et al., EMBO J. (1991) 10:2717-2723).
One aspect of the invention is directed to generating glycosylation site mutants in which the O- or N-linked glycosylation site of the binding protein has been mutated. One skilled in the art can generate such mutants using standard well-known technologies. Glycosylation site mutants that retain the biological activity, but have increased or decreased binding activity, are another object of the invention.
In still another embodiment, the glycosylation of the anti-B7-H3 antibody or antigen binding portion of the invention is modified. For example, an aglycoslated antibody can be made (i.e., the antibody lacks glycosylation). Glycosylation can be altered to, for example, increase the affinity of the antibody for antigen Such carbohydrate modifications can be accomplished by for example altering one or more sites of glycosylation within the antibody sequence. For example, one or more amino acid substitutions can be made that result in elimination of one or more variable region glycosylation sites to thereby eliminate glycosylation at that site. Such aglycosylation may increase the affinity of the antibody for antigen. Such an approach is described in further detail in PCT Publication WO2003016466A2, and U.S. Pat. Nos.5,714,350 and 6,350,861, each of which is incorporated herein by reference in its entirety.
Additionally or alternatively, a modified anti-B7-H3 antibody of the invention can be made that has an altered type of glycosylation, such as a hypofucosylated antibody having reduced amounts of fucosyl residues or an antibody having increased bisecting GlcNAc structures. Such altered glycosylation patterns have been demonstrated to increase the ADCC ability of antibodies. Such carbohydrate modifications can be accomplished by, for example, expressing the antibody in a host cell with altered glycosylation machinery. Cells with altered glycosylation machinery have been described in the art and can be used as host cells in which to express recombinant antibodies of the invention to thereby produce an antibody with altered glycosylation. See, for example, Shields, R. L. et al. (2002) J. Biol. Chem.277:26733-26740; Umana et al. (1999) Nat. Biotech.17:176-1, as well as, European Patent No: EP 1,176,195; PCT Publications WO 03/035835; WO 99/5434280, each of which is incorporated herein by reference in its entirety.
Protein glycosylation depends on the amino acid sequence of the protein of interest, as well as the host cell in which the protein is expressed. Different organisms may produce different glycosylation enzymes (e.g., glycosyltransferases and glycosidases), and have different substrates (nucleotide sugars) available. Due to such factors, protein glycosylation pattern, and composition of glycosyl residues, may differ depending on the host system in which the particular protein is expressed. Glycosyl residues useful in the invention may include, but are not limited to, glucose, galactose, mannose, fucose, n-acetylglucosamine and sialic acid. Preferably the glycosylated binding protein comprises glycosyl residues such that the glycosylation pattern is human.
Differing protein glycosylation may result in differing protein characteristics. For instance, the efficacy of a therapeutic protein produced in a microorganism host, such as yeast, and
glycosylated utilizing the yeast endogenous pathway may be reduced compared to that of the same protein expressed in a mammalian cell, such as a CHO cell line. Such glycoproteins may also be immunogenic in humans and show reduced half-life in vivo after administration. Specific receptors in humans and other animals may recognize specific glycosyl residues and promote the rapid clearance of the protein from the bloodstream. Other adverse effects may include changes in protein folding, solubility, susceptibility to proteases, trafficking, transport, compartmentalization, secretion, recognition by other proteins or factors, antigenicity, or allergenicity. Accordingly, a practitioner may prefer a therapeutic protein with a specific composition and pattern of glycosylation, for example glycosylation composition and pattern identical, or at least similar, to that produced in human cells or in the species-specific cells of the intended subject animal Expressing glycosylated proteins different from that of a host cell may be achieved by genetically modifying the host cell to express heterologous glycosylation enzymes. Using recombinant techniques, a practitioner may generate antibodies or antigen binding portions thereof exhibiting human protein glycosylation. For example, yeast strains have been genetically modified to express non-naturally occurring glycosylation enzymes such that glycosylated proteins
(glycoproteins) produced in these yeast strains exhibit protein glycosylation identical to that of animal cells, especially human cells (U.S. patent Publication Nos.20040018590 and 20020137134 and PCT publication WO2005100584 A2).
Antibodies may be produced by any of a number of techniques. For example, expression from host cells, wherein expression vector(s) encoding the heavy and light chains is (are) transfected into a host cell by standard techniques. The various forms of the term“transfection” are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE- dextran transfection and the like. Although it is possible to express antibodies in either prokaryotic or eukaryotic host cells, expression of antibodies in eukaryotic cells is preferable, and most preferable in mammalian host cells, because such eukaryotic cells (and in particular mammalian cells) are more likely than prokaryotic cells to assemble and secrete a properly folded and immunologically active antibody.
Preferred mammalian host cells for expressing the recombinant antibodies of the invention include Chinese Hamster Ovary (CHO cells) (including dhfr- CHO cells, described in Urlaub and Chasin, (1980) Proc. Natl. Acad. Sci. USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in R.J. Kaufman and P.A. Sharp (1982) Mol. Biol.159:601-621), NS0 myeloma cells, COS cells and SP2 cells. When recombinant expression vectors encoding antibody genes are introduced into mammalian host cells, the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or, more preferably, secretion of the antibody into the culture medium in which the host cells are grown. Antibodies can be recovered from the culture medium using standard protein purification methods.
Host cells can also be used to produce functional antibody fragments, such as Fab fragments or scFv molecules. It will be understood that variations on the above procedure are within the scope of the invention. For example, it may be desirable to transfect a host cell with DNA encoding functional fragments of either the light chain and/or the heavy chain of an antibody of this invention. Recombinant DNA technology may also be used to remove some, or all, of the DNA encoding either or both of the light and heavy chains that is not necessary for binding to the antigens of interest. The molecules expressed from such truncated DNA molecules are also encompassed by the antibodies of the invention. In addition, bifunctional antibodies may be produced in which one heavy and one light chain are an antibody of the invention and the other heavy and light chain are specific for an antigen other than the antigens of interest by crosslinking an antibody of the invention to a second antibody by standard chemical crosslinking methods.
In a preferred system for recombinant expression of an antibody, or antigen binding portion thereof, a recombinant expression vector encoding both the antibody heavy chain and the antibody light chain is introduced into dhfr- CHO cells by calcium phosphate-mediated transfection. Within the recombinant expression vector, the antibody heavy and light chain genes are each operatively linked to CMV enhancer/AdMLP promoter regulatory elements to drive high levels of transcription of the genes. The recombinant expression vector also carries a DHFR gene, which allows for selection of CHO cells that have been transfected with the vector using methotrexate selection/amplification. The selected transformant host cells are cultured to allow for expression of the antibody heavy and light chains and intact antibody is recovered from the culture medium. Standard molecular biology techniques are used to prepare the recombinant expression vector, transfect the host cells, select for transformants, culture the host cells and recover the antibody from the culture medium. Still further the invention provides a method of synthesizing a recombinant antibody of the invention by culturing a host cell in a suitable culture medium until a recombinant antibody is synthesized. Recombinant antibodies of the invention may be produced using nucleic acid molecules corresponding to the amino acid sequences disclosed herein The method can further comprise isolating the recombinant antibody from the culture medium.
The N- and C-termini of antibody polypeptide chains of the present invention may differ from the expected sequence due to commonly observed post-translational modifications. For example, C- terminal lysine residues are often missing from antibody heavy chains. Dick et al. (2008) Biotechnol. Bioeng.100:1132. N-terminal glutamine residues, and to a lesser extent glutamate residues, are frequently converted to pyroglutamate residues on both light and heavy chains of therapeutic antibodies. Dick et al. (2007) Biotechnol. Bioeng.97:544; Liu et al. (2011) JBC 28611211; Liu et al. (2011) J. Biol. Chem.286:11211. III. Anti-B7-H3 Antibody Drug Conjugates (ADCs)
Anti-B7-H3 antibodies described herein may be conjugated to a drug moiety to form an anti- B7-H3 Antibody Drug Conjugate (ADC). Antibody-drug conjugates (ADCs) may increase the therapeutic efficacy of antibodies in treating disease, e.g., cancer, due to the ability of the ADC to selectively deliver one or more drug moiety(s) to target tissues, such as a tumor-associated antigen, e.g., B7-H3 expressing tumors. Thus, in certain embodiments, the invention provides anti-B7-H3 ADCs for therapeutic use, e.g., treatment of cancer.
Anti-B7-H3 ADCs of the invention comprise an anti-B7-H3 antibody, i.e., an antibody that specifically binds to B7-H3, linked to one or more drug moieties. The specificity of the ADC is defined by the specificity of the antibody, i.e., anti-B7-H3. In one embodiment, an anti-B7-H3 antibody is linked to one or more cytotoxic drug(s) which is delivered internally to a transformed cancer cell expressing B7-H3.
Examples of drugs that may be used in the anti-B7-H3 ADC of the invention are provided below, as are linkers that may be used to conjugate the antibody and the one or more drug(s). The terms“drug,”“agent,” and“drug moiety” are used interchangeably herein. The terms“linked” and “conjugated” are also used interchangeably herein and indicate that the antibody and moiety are covalently linked.
In some embodiments, the ADC has the following formula (formula I):
Figure imgf000087_0001
wherein Ab is the antibody, e.g., anti-B7-H3 antibody huAb13v1, huAb3v2.5, or huAb3v2.6, and (L) is a linker, (D) is a drug, and LK represents a covalent linkage linking linker L to antibody Ab; and m is an integer ranging from 1 to 20. D is a drug moiety having, for example, cytostatic, cytotoxic, or otherwise therapeutic activity against a target cell, e.g., a cell expressing B7-H3. In some
embodiments, m ranges from 1 to 8, 1 to 7, 1 to 6, 2 to 6, 1 to 5, 1 to 4, 1 to 3, 1 to 2, 1.5 to 8, 1.5 to 7, 1.5 to 6, 1.5 to 5, 1.5 to 4, 2 to 6, 1 to 5, 1 to 4, 1 to 3, 1 to 2,or 2 to 4. The DAR of an ADC is equivalent to the“m” referred to in Formula I. In one embodiment, the ADC has a formula of Ab- (LK-L-D)m, wherein Ab is an anti-B7-H3 antibody, e.g.huAb13v1, huAb3v2.5, or huAb3v2.6, L is a linker, D is a drug, e.g., a Bcl-xL inhibitor, LK is a covalent linker, e.g., -S-, and m is 1 to 8 (or a DAR of 2-4). Additional details regarding drugs (D of Formula I) and linkers (L of Formula I) that may be used in the ADCs of the invention, as well as alternative ADC structures, are described below. III. A. Anti-B7-H3 ADCs: Bcl-xL Inhibitors, Linkers, Synthons, and Methods of Making Same Dysregulated apoptotic pathways have also been implicated in the pathology of cancer. The implication that down-regulated apoptosis (and more particularly the Bcl-2 family of proteins) is involved in the onset of cancerous malignancy has revealed a novel way of targeting this still elusive disease. Research has shown, for example, the anti-apoptotic proteins, Bcl 2 and Bcl-xL, are over- expressed in many cancer cell types. See, Zhang, 2002, Nature Reviews/Drug Discovery 1:101;
Kirkin et al., 2004, Biochimica Biophysica Acta 1644:229-249; and Amundson et al., 2000, Cancer Research 60:6101-6110. The effect of this deregulation is the survival of altered cells which would otherwise have undergone apoptosis in normal conditions. The repetition of these defects associated with unregulated proliferation is thought to be the starting point of cancerous evolution.
Aspects of the disclosure concern anti-B7-H3 ADCs comprising an anti-B7-H3 antibody conjugated to a drug via a linker, wherein the drug is a Bcl-xL inhibitor. In specific embodiments, the ADCs are compounds according to structural formula (I) below, or a pharmaceutically acceptable salt thereof, wherein Ab represents the anti-B7-H3 antibody, D represents a Bcl-xL inhibitor drug (i.e., a compound of formula IIa or IIb as shown below), L represents a linker, LK represents a covalent linkage linking the linker (L) to the anti-B7-H3 antibody (Ab) and m represents the number of D-L- LK units linked to the antibody, which is an integer ranging from 1 to 20. In certain embodiments, m is 2, 3 or 4. In some embodiments, m ranges from 1 to 8, 1 to 7, 1 to 6, 2 to 6, 1 to 5, 1 to 4, 1 to 3, 1 to 2, or 2 to 4.
Figure imgf000088_0001
Specific embodiments of various Bcl-xL inhibitors per se, and various Bcl-xL inhibitors (D), linkers (L) and anti-B7-H3 antibodies (Ab) that can comprise the ADCs described herein, as well as the number of Bcl-xL inhibitors linked to the ADCs, are described in more detail below.
Examples of Bcl-xL inhibitors that may be used in the anti-B7-H3 ADC of the invention are provided below, as are linkers that may be used to conjugate the antibody and the one or more Bcl-xL inhibitor(s). The terms“linked” and“conjugated” are also used interchangeably herein and indicate that the antibody and moiety are covalently linked. III.A.1. Bcl-xL Inhibitors
The Bcl-xL inhibitors may be used as compounds or salts per se in the various methods described herein, or may be included as a component part of an ADC, e.g., as the drug (D) in formula (I).
Specific embodiments of Bcl-xL inhibitors that may be used in unconjugated form, or that may be included as part of an ADC include compounds according to structural formula (IIa) or (IIb). In the present invention, when the Bcl-xL inhibitors are included as part of an ADC, # shown in formula (IIa) or (IIb) below represents a point of attachment to a linker, which indicates that they are represented in a monoradical form.
Figure imgf000088_0002
Figure imgf000089_0001
,
Figure imgf000089_0002
tly selected from halo, hydroxy, nitro, lower alkyl, lower heteroalkyl, C1-4alkoxy, amino, cyano and halomethyl;
,
Figure imgf000089_0003
independently selected from halo, hydroxy, nitro, lower alkyl, lower heteroalkyl, C1-4alkoxy, amino, cyano and halomethyl, wherein the #-N(R4)-R13-Z2b- substituent of formula (IIb) is attached to Ar2 at any Ar2 atom capable of being substituted;
Z1 is selected from N, CH, C-halo and C-CN;
Z2a, Z2b, and Z2c are each, independent from one another, selected from a bond, NR6, CR6aR6b, O, S, S(O), S(O)2, NR6C(O), NR6aC(O)NR6b, and NR6C(O)O; R1 is selected from hydrogen, methyl, halo, halomethyl, ethyl and cyano;
R2 is selected from hydrogen, methyl, halo, halomethyl and cyano;
R3 is selected from hydrogen, lower alkyl and lower heteroalkyl;
R4 is selected from hydrogen, lower alkyl, monocyclic cycloalkyl, monocyclic heterocyclyl, lower heteroalkyl or is taken together with an atom of R13 to form a cycloalkyl or heterocyclyl ring having between 3 and 7 ring atoms, wherein the lower alkyl, monocyclic cycloalkyl, monocyclic heterocyclyl, lower heteroalkyl are optionally substituted with one or more halo, cyano, C1-4alkoxy, monocyclic cycloalkyl, monocyclic heterocyclyl, NHC(O)CR6aR6b, NHS(O)CR6aR6b,
NHS(O)2CR6aR6b, S(O)2CR6aR6b or S(O)2NH2 groups;
R6, R6a and R6b are each, independent from one another, selected from hydrogen, lower alkyl, lower heteroalkyl, optionally substituted monocyclic cycloalklyl and monocyclic heterocyclyl, or are taken together with an atom from R13 to form a cycloalkyl or heterocyclyl ring having between 3 and 7 ring atoms;
R10 is selected from cyano, OR14, SR14, SOR14, SO2R14, SO2NR14aR14b, NR14aR14b, NHC(O)R14 and NHSO2R14;
R11a and R11b are each, independently of one another, selected from hydrogen, halo, methyl, ethyl, halomethyl, hydroxyl, methoxy, CN, and SCH3;
R12 is selected from hydrogen, halo, cyano, lower alkyl, lower heteroalkyl, cycloalkyl, or heterocyclyl, wherein the alkyl, heteroalkyl, cycloalkyl, or heterocyclyl are optionally substituted with one or more halo, cyano, C1-4alkoxy, monocyclic cycloalkyl, monocyclic heterocyclyl,
NHC(O)CR6aR6b, NHS(O)CR6aR6b, NHS(O)2CR6aR6b or S(O)2CR6aR6b groups;
R13 is selected from a bond, optionally substituted lower alkylene, optionally substituted lower heteroalkylene, optionally substituted cycloalkyl or optionally substituted heterocyclyl;
R14 is selected from hydrogen, optionally substituted lower alkyl and optionally substituted lower heteroalkyl;
R14a and R14b are each, independently of one another, selected from hydrogen, optionally substituted lower alkyl, optionally substituted lower heteroalkyl, or are taken together with the nitrogen atom to which they are bonded to form a monocyclic cycloalkyl or monocyclic heterocyclyl ring;
R15 is selected from hydrogen, halo, C1-6 alkanyl, C2-4 alkenyl, C2-4 alkynyl, and C1-4 haloalkyl and C1-4 hydroxyalkyl, with the proviso that when R15 is present, R4 is not C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C1-4 haloalkyl or C1-4 hydroxyalkyl, wherein the R4 C1-6 alkanyl, C2-4 alkenyl, C2-4 alkynyl, C1- 4 haloalkyl and C1-4 hydroxyalkyl are optionally substituted with one or more substituents
independently selected from OCH3, OCH2CH2OCH3, and OCH2CH2NHCH3; and
# represents a point of attachment to a linker or a hydrogen atom.
Specific embodiments of Bcl-xL inhibitors that may be used in unconjugated form, or that may be included as part of an ADC include compounds according to structural formula (IIa) or (IIb): (IIa)
Figure imgf000091_0001
(IIb)
Figure imgf000091_0002
r l hr f hrin
Figure imgf000091_0003
selected from halo, hydroxy, nitro, lower alkyl, lower heteroalkyl, C1-4alkoxy, amino, cyano and halomethyl;
,
Figure imgf000091_0004
Figure imgf000092_0001
Figure imgf000092_0002
is optionally substituted with one or more substituents independently selected from halo, hydroxy, nitro, lower alkyl, lower heteroalkyl, C1-4alkoxy, amino, cyano and halomethyl, wherein the #-N(R4)-R13-Z2b- substituent of formula (IIb) is attached to Ar2 at any Ar2 atom capable of being substituted;
Z1 is selected from N, CH, C-halo and C-CN;
Z2a, Z2b, and Z2c are each, independent from one another, selected from a bond, NR6, CR6aR6b, O, S, S(O), S(O)2, NR6C(O), NR6aC(O)NR6b, and NR6C(O)O;
R1 is selected from hydrogen, methyl, halo, halomethyl, ethyl and cyano;
R2 is selected from hydrogen, methyl, halo, halomethyl and cyano;
R3 is selected from hydrogen, lower alkyl and lower heteroalkyl;
R4 is selected from hydrogen, lower alkyl, monocyclic cycloalkyl, monocyclic heterocyclyl, and lower heteroalkyl or is taken together with an atom of R13 to form a cycloalkyl or heterocyclyl ring having between 3 and 7 ring atoms, wherein the lower alkyl, monocyclic cycloalkyl, monocyclic heterocyclyl, and lower heteroalkyl are optionally substituted with one or more halo, cyano, hydroxy, C1-4alkoxy, monocyclic cycloalkyl, monocyclic heterocyclyl, C(O)NR6aR6b, S(O)2NR6aR6b,
NHC(O)CHR6aR6b, NHS(O)CHR6aR6b, NHS(O)2CHR6aR6b, S(O)2CHR6aR6b or S(O)2NH2 groups;
R6, R6a and R6b are each, independent from one another, selected from hydrogen, lower alkyl, lower heteroalkyl, optionally substituted monocyclic cycloalklyl and monocyclic heterocyclyl, or are taken together with an atom from R13 to form a cycloalkyl or heterocyclyl ring having between 3 and 7 ring atoms;
R10 is selected from cyano, OR14, SR14, SOR14, SO2R14, SO2NR14aR14b, NR14aR14b, NHC(O)R14 and NHSO2R14;
R11a and R11b are each, independently of one another, selected from hydrogen, halo, methyl, ethyl, halomethyl, hydroxyl, methoxy, CN, and SCH3;
R12 is selected from hydrogen, halo, cyano, lower alkyl, lower heteroalkyl, cycloalkyl, and heterocyclyl, wherein the alkyl, heteroalkyl, cycloalkyl, and heterocyclyl are optionally substituted with one or more halo, cyano, C1-4alkoxy, monocyclic cycloalkyl, monocyclic heterocyclyl,
NHC(O)CHR6aR6b, NHS(O)CHR6aR6b, NHS(O)2CHR6aR6b or S(O)2CHR6aR6b groups;
R13 is selected from a bond, optionally substituted lower alkylene, optionally substituted lower heteroalkylene, optionally substituted cycloalkyl or optionally substituted heterocyclyl; R14 is selected from hydrogen, optionally substituted lower alkyl and optionally substituted lower heteroalkyl;
R14a and R14b are each, independently of one another, selected from hydrogen, optionally substituted lower alkyl, and optionally substituted lower heteroalkyl, or are taken together with the nitrogen atom to which they are bonded to form an optionally substituted monocyclic cycloalkyl or monocyclic heterocyclyl ring;
R15 is selected from hydrogen, halo, C1-6 alkanyl, C2-4 alkenyl, C2-4 alkynyl, and C1-4 haloalkyl and C1-4 hydroxyalkyl, with the proviso that when R15 is present, R4 is not C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C1-4 haloalkyl or C1-4 hydroxyalkyl, wherein the R4 C1-6 alkanyl, C2-4 alkenyl, C2-4 alkynyl, C1- 4 haloalkyl and C1-4 hydroxyalkyl are optionally substituted with one or more substituents
independently selected from OCH3, OCH2CH2OCH3, and OCH2CH2NHCH3; and
# represents a point of attachment to a linker or a hydrogen atom.
Another embodiment of Bcl-xL inhibitors that may be used in unconjugated form, or that may be included as part of an ADC include compounds according to structural formula (IIa) or (IIb):
Figure imgf000093_0001
Figure imgf000093_0002
or salts thereof, wherein: Ar1 is selected from
Figure imgf000094_0001
,
Figure imgf000094_0002
and is optionally substituted with one or more substituents independently selected from halo, hydroxy, nitro, lower alkyl, lower heteroalkyl, C1-4alkoxy, amino, cyano and halomethyl;
,
Figure imgf000094_0003
Figure imgf000094_0004
and is optionally substituted with one or more substituents independently selected from halo, hydroxy, nitro, lower alkyl, lower heteroalkyl, C1- 4alkoxy, amino, cyano and halomethyl, wherein the #-N(R4)-R13-Z2b- substituent of formula (IIb) is attached to Ar2 at any Ar2 atom capable of being substituted;
Z1 is selected from N, CH, C-halo and C-CN;
Z2a, Z2b, and Z2c are each, independent from one another, selected from a bond, NR6, CR6aR6b, O, S, S(O), SO2, NR6C(O), NR6aC(O)NR6b, and NR6C(O)O;
R1 is selected from hydrogen, methyl, halo, halomethyl, ethyl and cyano;
R2 is selected from hydrogen, methyl, halo, halomethyl and cyano;
R3 is selected from hydrogen, lower alkyl and lower heteroalkyl;
R4 is selected from hydrogen, lower alkyl, monocyclic cycloalkyl, monocyclic heterocyclyl, lower heteroalkyl or is taken together with an atom of R13 to form a cycloalkyl or heterocyclyl ring having between 3 and 7 ring atoms, wherein the lower alkyl, monocyclic cycloalkyl, monocyclic heterocyclyl, lower heteroalkyl are optionally substituted with one or more halo, cyano, C1-4alkoxy, monocyclic cycloalkyl, monocyclic heterocyclyl, NC(O)CR6aR6b, NS(O)CR6aR6b, NS(O) 6a
2CR R6b, S(O)2CR6aR6b or S(O)2NH2 groups; R6, R6a and R6b are each, independent from one another, selected from hydrogen, lower alkyl, lower heteroalkyl, optionally substituted monocyclic cycloalklyl and monocyclic heterocyclyl, or are taken together with an atom from R13 to form a cycloalkyl or heterocyclyl ring having between 3 and 7 ring atoms;
R10 is selected from cyano, OR14, SR14, SOR14, SO2R14, SO2NR14aR14b, NR14aR14b, NHC(O)R14 and NHSO2R14;
R11a and R11b are each, independently of one another, selected from hydrogen, halo, methyl, ethyl, halomethyl, hydroxyl, methoxy, CN, and SCH3;
R12 is selected from hydrogen, halo, cyano, lower alkyl, lower heteroalkyl, cycloalkyl, or heterocyclyl, wherein the alkyl, heteroalkyl, cycloalkyl, or heterocyclyl are optionally substituted with one or more halo, cyano, C1-4alkoxy, monocyclic cycloalkyl, monocyclic heterocyclyl,
NHC(O)CR6aR6b, NHS(O)CR6aR6b, NHS(O)2CR6aR6b or S(O)2CR6aR6b groups;
R13 is selected from a bond, optionally substituted lower alkylene, optionally substituted lower heteroalkylene, optionally substituted cycloalkyl or optionally substituted heterocyclyl;
R14 is selected from hydrogen, optionally substituted lower alkyl and optionally substituted lower heteroalkyl;
R14a and R14b are each, independently of one another, selected from hydrogen, optionally substituted lower alkyl, optionally substituted lower heteroalkyl, or are taken together with the nitrogen atom to which they are bonded to form a monocyclic cycloalkyl or monocyclic heterocyclyl ring;
R15 is selected from hydrogen, halo, C1-6 alkanyl, C2-4 alkenyl, C2-4 alkynyl, and C1-4 haloalkyl and C1-4 hydroxyalkyl, with the proviso that when R15 is present, R4 is not C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C1-4 haloalkyl or C1-4 hydroxyalkyl, wherein the R4 C1-6 alkanyl, C2-4 alkenyl, C2-4 alkynyl, C1- 4 haloalkyl and C1-4 hydroxyalkyl are optionally substituted with one or more substituents
independently selected from OCH3, OCH2CH2OCH3, and OCH2CH2NHCH3; and
# represents a point of attachment to a linker or a hydrogen atom.
When a Bcl-xL inhibitor of structural formulae (IIa) and (IIb) is not a component of an ADC, # in formulae (IIa) and (IIb) represents the point of attachment to a hydrogen atom. When the Bcl-xL inhibitor is a component of an ADC, # in formulae (IIa) and (IIb) represents the point of attachment to a the linker. When a Bcl-xL inhibitor is a component of an ADC, the ADC may comprise one or more Bcl-xL inhibitors, which may be the same or different, but are typically the same. In certain embodiments, Ar1 of formula (IIa) or (IIb) is selected from
Figure imgf000096_0001
, and
Figure imgf000096_0002
and is optionally substituted with one or more substituents independently selected from halo,
cyano, methyl, and halomethyl. In particular embodiments, Ar1 is
Figure imgf000096_0003
. In particular
embodiments, Ar1 is unsubstituted.
In all embodiments, the #-N(R4)-R13-Z2b- substituent of formula (IIb) is attached to Ar2 at any Ar2 atom capable of being substituted.
In certain embodiments, Ar2 of formula (IIa) or (IIb) is
Figure imgf000096_0004
which is optionally substituted at the 5- osition with a rou selected from h drox l C1-4 alkox and cyano; or
In certa .
In certa
Figure imgf000096_0005
. In certain embodiments, Ar2 of formula (IIa) or (IIb) is In certain embodiments, Ar2 of formula (IIa) or (IIb) is In certain embodiments, Ar2 of formula (IIa) or (IIb) is . In certain embodiments, Ar2 of formula (IIa) or (IIb) is In certain embodiments, Ar2 of formula (IIa) or (IIb) is
In certain embodiments, Ar2 of formula (IIa) or (IIb) is
In certain embodiments, Ar2 of formula (IIa) or (IIb) is In certain embodiments, Ar2 of formula (IIa) or (IIb) is In certain embodiments, Ar2 of formula (IIa) or (IIb) is
In certain embodiments, Ar2 of formula (IIa) or (IIb) is In certain embodiments, Ar2 of formula (IIa) or (IIb) is
Figure imgf000097_0001
In certain embodiments, Ar2 of formula (IIa) or (IIb) is
In certain embodiments, Ar2 of formula (IIa) or (IIb) is
In certain embodiments, Ar2 of formula (IIa) or (IIb) is In certain embodiments, Ar2 of formula (IIa) or (IIb) is In certain embodiments, Ar2 of formula (IIa) or (IIb) is . In certain embodiments, Ar2 of formula (IIa) or (IIb) is
In certain embodiments, Ar2 of formula (IIa) or (IIb) is In certain embodiments, Ar2 of formula (IIa) or (IIb) is In certain embodiments, Ar2 of formula (IIa) or (IIb) is In certain embodiments, Ar2 of formula (IIa) or (IIb) is
Figure imgf000098_0001
In certain embodiments, Ar2 of formula (IIa) or (IIb) is
In certain embodiments, Ar2 of formula (IIa) or (IIb) is . In certain embodiments, Ar2 of formula (IIa) is unsubstituted.
In certain embodiments, Ar2 of formula (IIa) or (IIb) is
Figure imgf000099_0001
, which is substituted at the 5-position with a group selected from hydroxyl, C1-4alkoxy, and cyano.
In certain embodiments, Z1 of formula (IIa) or (IIb) is N.
In certain embodiments, R1 of formula (IIa) or (IIb) is selected from methyl and chloro.
In certain embodiments, R2 of formula (IIa) or (IIb) is selected from hydrogen and methyl. In particular embodiments, R2 is hydrogen.
In certain embodiments, R4 of formula (IIa) or (IIb) is methyl.
In certain embodiments, R4 of formula (IIa) or (IIb) is (CH2)2OCH3.
In certain embodiments, R4 of formula (IIa) or (IIb) is hydrogen.
In certain embodiments, R4 of formula (IIa) or (IIb) is monocyclic heterocyclyl, wherein the monocyclic heterocycloalkyl is substituted with one S(O)2CH3.
In certain embodiments, R4 of formula (IIa) or (IIb) is hydrogen or lower alkyl, wherein the lower alkyl is optionally substituted with C1-4 alkoxy or C(O)NR6aR6b.
In certain embodiments, R4 of formula (IIa) or (IIb) is lower alkyl, wherein the lower alkyl is substituted with C(O)NH2.
In certain embodiments, R4 of formula (IIa) or (IIb) is lower alkyl, wherein the lower alkyl is substituted with S(O)2NH2.
In certain embodiments, R4 of formula (IIa) or (IIb) is lower alkyl, wherein the lower alkyl is substituted with hydroxy.
In certain embodiments, R4 of formula (IIa) or (IIb) is lower alkyl, wherein the lower alkyl is substituted with C(O)N(CH3)2.
In certain embodiments, R4 of formula (IIa) or (IIb) is lower alkyl, wherein the lower alkyl is substituted with C(O)NHCH3.
In certain embodiments, R11a and R11b of formula (IIa) or (IIb) are the same. In a particular embodiment, R11a and R11b are each methyl. In another embodiment, R11a and R11b are each ethyl. In another embodiment, R11a and R11b are each methoxy. In certain embodiments, R11a and R11b of formula (IIa) or (IIb) are independently selected from F, Br and Cl.
In certain embodiments, Z1 is N, Z2a is O, R1 is methyl or chloro, R2 is hydrogen, and Ar2 is
Figure imgf000100_0001
is optionally substituted at the 5-position with a group selected from hydroxyl, C1-4 alkoxy, and cyano.
Certain embodiments pertain to a compound of formula (IIa). In certain embodiments, Z2a of formula (IIa) is O.
In certain embodiments, Z2a of formula (IIa) is CH2 or O.
In certain embodiments, Z2a of formula (IIa) is S.
In certain embodiments, Z2a of formula (IIa) is CH2.
In certain embodiments, Z2a of formula (IIa) is NR6. In some such embodiments R6 is methyl. In certain embodiments, Z2a of formula (IIa) is NR6C(O). In some such embodiments R6 is hydrogen.
In certain embodiments, Z2a of formula (IIa) is O, R13 is ethylene, and R4 is lower alkyl. In certain embodiments, Z2a of formula (IIa) is O, R13 is ethylene, and R4 is hydrogen or lower alkyl optionally substituted with C1-4 alkoxy or C(O)NR6aR6b.
In certain embodiments, Z2a of formula (IIa) is O, R13 is ethylene, and R4 is methyl.
In certain embodiments, Z2a of formula (IIa) is O, R13 is ethylene, and R4 is hydrogen.
In certain embodiments, Z2a of formula (IIa) is NR6C(O), R6 is hydrogen, R13 is methylene, and R4 is hydrogen.
In certain embodiments, Z2a of formula (IIa) is S, R13 is ethylene, and R4 is hydrogen.
In certain embodiments, Z2a of formula (IIa) is CH2, R13 is ethylene, and R4 is hydrogen. In certain embodiments, the group R13 in formula (IIa) is ethylene. In some such
embodiments Z2a is O.
In certain embodiments, the group R13 in formula (IIa) is propylene. In some such embodiments Z2a is O.
In certain embodiments, the group R13 in formula (IIa) is selected from lower alkylene or lower heteroalkylene.
In certain embodiments, the group R13 in formula (IIa) is selected from (CH2)2O(CH2)2, (CH2)3O(CH2)2, (CH2)2O(CH2)3 and (CH2)3O(CH2)3. In some such embodiments Z2a is O.
In certain embodiments, the group R13 in formula (IIa) is selected from (CH2)2(SO2)(CH2)2, (CH2)3(SO2)(CH2)2, (CH2)2(SO2)(CH2)3 and (CH2)3(SO2)(CH2)3. In some such embodiments Z2a is O.
In certain embodiments, the group R13 in formula (IIa) is selected from (CH2)2(SO)(CH2)2, (CH2)2(SO)(CH2)3, (CH2)3(SO)(CH2)2 and (CH2)3(SO)(CH2)3. In some such embodiments Z2a is O. In certain embodiments, the group R13 in formula (IIa) is selected from (CH2)2S(CH2)2, (CH2)2S(CH2)3, (CH2)3S(CH2)2 and (CH2)3S(CH2)3. In some such embodiments Z2a is O.
in formula (IIa) is
Figure imgf000101_0001
in formula (IIa) is
Figure imgf000101_0002
n certan emo ments, te group n ormua a s in formula (IIa) is
Figure imgf000101_0003
In r in m imn h roup
Figure imgf000101_0004
Figure imgf000101_0006
Figure imgf000101_0005
and . .
Figure imgf000102_0001
ceta e bod ets, te go
Figure imgf000102_0002
.
In r in m imn h r
Figure imgf000102_0003
in formula (IIa) is
Figure imgf000102_0004
in formula (IIa) is
Figure imgf000102_0005
Figure imgf000103_0001
p p ( ) In certain embodiments, the group Z2b in formula (IIb) is a bond, O, or NR6, or and R13 is ethylene or optionally substituted heterocyclyl.
In certain embodiments, the group Z2b in formula (IIb) is NR6. In some such embodiments R6 is methyl.
In certain embodiments, the group Z2b in formula (IIb) is NR6 and R13 is ethylene. In some such embodiments R6 is methyl.
In certain embodiments, the group Z2b in formula (IIb) is O and R13 is ethylene. In some such embodiments R4 is methyl.
In certain embodiments, the group Z2b in formula (IIb) is NR6, wherein the R6 group is taken together with an atom of R13 to form a ring having between 4 and 6 atoms. In some such
embodiments the ring is a five membered ring.
In certain embodiments, the group Z2b in formula (IIb) is methylene and the group R13 is methylene.
In certain embodiments, the group Z2b in formula (IIb) is methylene and the group R13 is a bond.
In certain embodiments, the group Z2b in formula (IIb) is oxygen and the group R13 is selected from (CH2)2O(CH2)2, (CH2)3O(CH2)2, (CH2)2O(CH2)3 and (CH2)3O(CH2)3. In some such
embodiments R4 is methyl.
In certain embodiments, the group Z2c in formula (IIb) is a bond and R12 is OH.
In certain embodiments, the group Z2c in formula (IIb) is a bond and R12 is selected from F, Cl, Br and I.
In certain embodiments, the group Z2c in formula (IIb) is a bond and R12 is lower alkyl. In some such embodiments R12 is methyl.
In certain embodiments, the group Z2c in formula (IIb) is O and R12 is a lower heteroalkyl. In some such embodiments R12 is O(CH2)2OCH3.
In certain embodiments, the group Z2c in formula (IIb) is O and R12 is lower alkyl optionally substituted with one or more halo or C1-4 alkoxy.
In certain embodiments, the group Z2c in formula (IIb) is O and R12 is a lower alkyl. In particular embodiments R12 is methyl.
In certain embodiments, the group Z2c in formula (IIb) is S and R12 is a lower alkyl. In some such embodiments R12 is methyl.
Exemplary Bcl-xL inhibitors according to structural formulae (IIa)-(IIb) that may be used in the methods described herein in unconjugated form and/or included in the ADCs described herein include the following compounds, and/or a pharmaceutically acceptable salt thereof:
Figure imgf000105_0001
Figure imgf000106_0001
Notably, when the Bcl-xL inhibitor of the present application is in conjugated form, the hydrogen corresponding to the # position of structural formula (IIa) or (IIb) is not present, forming a monoradical. For example, compound W3.01 (Example 1.1) is 6-[1-(1,3-benzothiazol-2- ylcarbamoyl)-1,2,3,4-tetrahydroquinolin-7-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid. When it is in uncon u ated form it has the followin structure:
Figure imgf000107_0001
When the same compound is included in the ADCs as shown in structural formula (IIa) or (IIb), the hydrogen corres ondin to the # osition is not resent formin a monoradical.
In certain embodiments, the Bcl-xL inhibitor is selected from the group consisting of W3.01, W3.02, W3.03, W3.04, W3.05, W3.06, W3.07, W3.08, W3.09, W3.10, W3.11, W3.12, W3.13, W3.14, W3.15, W3.16, W3.17, W3.18, W3.19, W3.20, W3.21, W3.22, W3.23, W3.24, W3.25, W3.26, W3.27, W3.28, W3.29, W3.30, W3.31, W3.32, W3.33, W3.34, W3.35, W3.36, W3.37, W3.38, W3.39, W3.40, W3.41, W3.42, W3.43, and pharmaceutically acceptable salts thereof (see Example 1 for compounds).
In certain embodiments, the ADC, or a pharmaceutically acceptable salt thereof, comprises a drug linked to an antibody by way of a linker, wherein the drug is a Bcl-xL inhibitor selected from the group consisting of W3.01, W3.02, W3.03, W3.04, W3.05, W3.06, W3.07, W3.08, W3.09, W3.10, W3.11, W3.12, W3.13, W3.14, W3.15, W3.16, W3.17, W3.18, W3.19, W3.20, W3.21, W3.22, W3.23, W3.24, W3.25, W3.26, W3.27, W3.28, W3.29, W3.30, W3.31, W3.32, W3.33, W3.34, W3.35, W3.36, W3.37, W3.38, W3.39, W3.40, W3.41, W3.42, W3.43. In certain embodiments, the ADC, or a pharmaceutically acceptable salt thereof, the Bcl-xL inhibitor is selected from the group consisting of the following compounds modified in that the hydrogen corresponding to the # position of structural formula (IIa) or (IIb) is not present forming a monoradical:
6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4-tetrahydroquinolin-7-yl]-3-[1-({3,5-dimethyl- 7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid;
6-[4-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-2H-1,4-benzoxazin-6-yl]-3-[1-({3,5- dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4- yl]pyridine-2-carboxylic acid;
6-[4-(1,3-benzothiazol-2-ylcarbamoyl)-1-methyl-1,2,3,4-tetrahydroquinoxalin-6-yl]-3-[1- ({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol- 4-yl]pyridine-2-carboxylic acid;
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-5,6-dihydroimidazo[1,5-a]pyrazin-7(8H)- yl]pyridine-2-carboxylic acid;
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-hydroxy-3,4-dihydroisoquinolin-2(1H)- yl]pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid;
3-[1-({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl- 1H-pyrazol-4-yl]-6-[8-([1,3]thiazolo[5,4-b]pyridin-2-ylcarbamoyl)naphthalen-2-yl]pyridine-2- carboxylic acid;
3-[1-({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl- 1H-pyrazol-4-yl]-6-[8-([1,3]thiazolo[4,5-b]pyridin-2-ylcarbamoyl)naphthalen-2-yl]pyridine-2- carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-3-[1- ({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol- 4-yl]pyridine-2-carboxylic acid;
6-[5-(1,3-benzothiazol-2-ylcarbamoyl)quinolin-3-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid;
6-[4-(1,3-benzothiazol-2-ylcarbamoyl)quinolin-6-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid; 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-3-{1- [(3-{2-[(2-methoxyethyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl- 1H-pyrazol-4-yl}pyridine-2-carboxylic acid;
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-cyano-3,4-dihydroisoquinolin-2(1H)- yl]pyridine-2-carboxylic acid;
6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4-tetrahydroquinolin-7-yl]-3-{1-[(3-{2-[(2- methoxyethyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol- 4-yl}pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-3-{1-[(3-{2-[(2- methoxyethyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol- 4-yl}pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-[1-({3,5- dimethyl-7-[2-(oxetan-3-ylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4- yl]pyridine-2-carboxylic acid;
6-[6-(3-aminopyrrolidin-1-yl)-8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl]-3-(1-{[3-(2-methoxyethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-{1-[(3,5- dimethyl-7-{2-[(2-sulfamoylethyl)amino]ethoxy}tricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H- pyrazol-4-yl}pyridine-2-carboxylic acid;
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-[3-(1,3-benzothiazol-2-ylcarbamoyl)-6,7-dihydrothieno[3,2-c]pyridin-5(4H)- yl]pyridine-2-carboxylic acid;
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-3-(trifluoromethyl)-5,6-dihydroimidazo[1,5- a]pyrazin-7(8H)-yl]pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-6-{methyl[2-(methylamino)ethyl]amino}-3,4- dihydroisoquinolin-2(1H)-yl]-3-(1-{[3-(2-methoxyethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-6-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-3-[1- ({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol- 4-yl]pyridine-2-carboxylic acid;
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-[4-(1,3-benzothiazol-2-ylcarbamoyl)quinolin-6-yl]pyridine-2-carboxylic acid; 6-[5-amino-8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-[1-({3,5- dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4- yl]pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-6-[3-(methylamino)prop-1-yn-1-yl]-3,4- dihydroisoquinolin-2(1H)-yl]-3-(1-{[3-(2-methoxyethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid;
6-[4-(1,3-benzothiazol-2-ylcarbamoyl)isoquinolin-6-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid;
6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-1H-indol-2-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid;
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-1H-indol-2-yl]pyridine-2-carboxylic acid;
6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-3-methyl-1H-indol-2-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-(1-{[3,5- dimethyl-7-(2-{[1-(methylsulfonyl)piperidin-4-yl]amino}ethoxy)tricyclo[3.3.1.13,7]dec-1-yl]methyl}- 5-methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-(1-{[3,5- dimethyl-7-(2-{[1-(methylsulfonyl)azetidin-3-yl]amino}ethoxy)tricyclo[3.3.1.13,7]dec-1-yl]methyl}- 5-methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid;
3-{1-[(3-{2-[(3-amino-3-oxopropyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl)methyl]-5-methyl-1H-pyrazol-4-yl}-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]pyridine-2-carboxylic acid;
6-[3-(1,3-benzothiazol-2-ylcarbamoyl)-1H-indazol-5-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid;
6-[3-(1,3-benzothiazol-2-ylcarbamoyl)-1H-indol-5-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid;
6-[3-(1,3-benzothiazol-2-ylcarbamoyl)-1H-pyrrolo[2,3-b]pyridin-5-yl]-3-[1-({3,5-dimethyl-7- [2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid; 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-((2-(N,N- dimethylsulfamoyl)ethyl)amino)ethoxy)-5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol- 4-yl)picolinic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-3-{1-[(3-{2-[(3- hydroxypropyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol- 4-yl}pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-(1-{[3-(2-{[3- (dimethylamino)-3-oxopropyl]amino}ethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5- methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-(1-{[3,5- dimethyl-7-(2-{[3-(methylamino)-3-oxopropyl]amino}ethoxy)tricyclo[3.3.1.13,7]dec-1-yl]methyl}-5- methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid;
3-(1-{[3-(2-aminoacetamido)-5,7-dimethyltricyclo[3.3.1.13,7]decan-1-yl]methyl}-5-methyl- 1H-pyrazol-4-yl)-6-{8-[(1,3-benzothiazol-2-yl)carbamoyl]-3,4-dihydroisoquinolin-2(1H)-yl}pyridine- 2-carboxylic acid;
3-[1-({3-[(2-aminoethyl)sulfanyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl- 1H-pyrazol-4-yl]-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]pyridine-2- carboxylic acid;
3-(1-{[3-(3-aminopropyl)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]pyridine-2- carboxylic acid; and
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]decan-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-{5-[(1,3-benzothiazol-2-yl)carbamoyl]quinolin-3-yl}pyridine-2-carboxylic acid.
The Bcl-xL inhibitors bind to and inhibit anti-apoptotic Bcl-xL proteins, inducing apoptosis. The ability of specific Bcl-xL inhibitors according to structural formulae (IIa)-(IIb) to bind to and inhibit Bcl-xL activity may be confirmed in standard binding and activity assays, including, for example, the TR-FRET Bcl-xL binding assays described in Tao et al., 2014, ACS Med. Chem. Lett., 5:1088-1093. A specific TR-FRET Bcl-xL binding assay that can be used to confirm Bcl-xL binding is provided in Example 4, below. Typically, Bcl-xL inhibitors useful as inhibitors per se and in the ADCs described herein will exhibit a Ki in the binding assay of Example 5 of less than about 1 nM, but may exhibit a significantly lower Ki, for example a Ki of less than about 1, 0.1, or even 0.01.
Bcl-xL inhibitory activity may also be confirmed in standard cell-based cytotoxicity assays, such as the FL5.12 cellular and Molt-4 cytotoxicity assays described in Tao et al., 2014, ACS Med. Chem. Lett., 5:1088-1093. A specific Molt-4 cellular cytotoxicity assay that may be used to confirm Bcl-xL inhibitory activity of specific Bcl-xL inhibitors that are able to permeate cell membranes is provided in Example 5, below. Typically, such cell-permeable Bcl-xL inhibitors will exhibit an EC50 of less than about 500 nM in the Molt-4 cytotoxicity assay of Example 5, but may exhibit a significantly lower EC50, for example an EC50 of less than about 250, 100, 50, 20, 10 or even 5 nM.
The process of mitochondrial outer-membrane permeabilization (MOMP) is controlled by the Bcl-2 family proteins. Specifically, MOMP is promoted by the pro-apoptotic Bcl-2 family proteins Bax and Bak which, upon activation oligomerize on the outer mitochondrial membrane and form pores, leading to release of cytochrome c (cyt c). The release of cyt c triggers formulation of the apoptosome which, in turn, results in caspase activation and other events that commit the cell to undergo programmed cell death (see, Goldstein et al., 2005, Cell Death and Differentiation 12:453- 462). The oligomerization action of Bax and Bak is antagonized by the anti-apoptotic Bcl-2 family members, including Bcl-2 and Bcl-xL. Bcl-xL inhibitors, in cells that depend upon Bcl-xL for survival, can cause activation of Bax and/or Bak, MOMP, release of cyt c and downstream events leading to apoptosis. The process of cyt c release can be assessed via western blot of both mitochondrial and cytosolic fractions of cytochrome c in cells and used as a proxy measurement of apoptosis in cells.
As a means of detecting Bcl-xL inhibitory activity and consequent release of cyt c, the cells can be treated with an agent that causes selective pore formation in the plasma, but not mitochondrial, membrane. Specifically, the cholesterol/phospholipid ratio is much higher in the plasma membrane than the mitochondrial membrane. As a result, short incubation with low concentrations of the cholesterol-directed detergent digitonin selectively permeabilizes the plasma membrane without significantly affecting the mitochondrial membrane. This agent forms insoluble complexes with cholesterol leading to the segregation of cholesterol from its normal phospholipid binding sites. This action, in turn, leads to the formation of holes about 40-50 Å wide in the lipid bilayer. Once the plasma membrane is permeabilized, cytosolic components able to pass over digitonin-formed holes can be washed out, including the cytochrome C that was released from mitochondria to cytosol in the apoptotic cells (Campos, 2006, Cytometry A 69(6):515-523).
Although many of the Bcl-xL inhibitors of structural formulae (IIa)-(IIb) selectively or specifically inhibit Bcl-xL over other anti-apoptotic Bcl-2 family proteins, selective and/or specific inhibition of Bcl-xL is not necessary. The Bcl-xL inhibitors and ADCs comprising the compounds may also, in addition to inhibiting Bcl-xL, inhibit one or more other anti-apoptotic Bcl-2 family proteins, such as, for example, Bcl-2. In some embodiments, the Bcl-xL inhibitors and/or ADCs are selective and/or specific for Bcl-xL. By specific or selective is meant that the particular Bcl-xL inhibitor and/or ADC binds or inhibits Bcl-xL to a greater extent than Bcl-2 under equivalent assay conditions. In specific embodiments, the Bcl-xL inhibitors and/or ADCs exhibit in the range of about 10-fold, 100-fold, or even greater specificity or selectivity for Bcl-xL than Bcl-2 in binding assays. III.A.2. Bcl-xL Linkers
In the ADCs described herein, the Bcl-xL inhibitors (described in Section III.A) are linked to the anti-B7-H3 antibody by way of linkers. The linker linking a Bcl-xL inhibitor to the anti-B7-H3 antibody of an ADC may be short, long, hydrophobic, hydrophilic, flexible or rigid, or may be composed of segments that each independently has one or more of the above-mentioned properties such that the linker may include segments having different properties. The linkers may be polyvalent such that they covalently link more than one Bcl-xL inhibitor to a single site on the antibody, or monovalent such that covalently they link a single Bcl-xL inhibitor to a single site on the antibody.
As will be appreciated by skilled artisans, the linkers link the Bcl-xL inhibitors to the anti- B7-H3 antibody by forming a covalent linkage to the Bcl-xL inhibitor at one location and a covalent linkage to antibody at another. The covalent linkages are formed by reaction between functional groups on the linker and functional groups on the inhibitors and antibody. As used herein, the expression“linker” is intended to include (i) unconjugated forms of the linker that include a functional group capable of covalently linking the linker to a Bcl-xL inhibitor and a functional group capable of covalently linking the linker to an anti-B7-H3 antibody; (ii) partially conjugated forms of the linker that include a functional group capable of covalently linking the linker to an anti-B7-H3 antibody and that is covalently linked to a Bcl-xL inhibitor, or vice versa; and (iii) fully conjugated forms of the linker that is covalently linked to both a Bcl-xL inhibitor and an anti-B7-H3 antibody. In some specific embodiments of intermediate synthons and ADCs described herein, moieties comprising the functional groups on the linker and covalent linkages formed between the linker and antibody are specifically illustrated as Rx and LK, respectively. One embodiment pertains to an ADC formed by contacting an antibody that binds a cell surface receptor or tumor associated antigen expressed on a tumor cell with a synthon described herein under conditions in which the synthon covalently links to the anti-B7-H3 antibody. One embodiment pertains to a method of making an ADC formed by contacting a synthon described herein under conditions in which the synthon covalently links to the anti-B7-H3 antibody. One embodiment pertains to a method of inhibiting Bcl-xL activity in a cell that expresses Bcl-xL, comprising contacting the cell with an ADC described herein that is capable of binding the cell, under conditions in which the ADC binds the cell.
Exemplary polyvalent linkers that may be used to link many Bcl-xL inhibitors to an antibody are described, for example, in U.S. Patent No 8,399,512; U.S. Published Application No.
2010/0152725; U.S. Patent No.8,524,214; U.S. Patent No.8,349,308; U.S. Published Application No. 2013/189218; U.S. Published Application No. 2014/017265; WO 2014/093379; WO 2014/093394; WO 2014/093640, the contents of which are incorporated herein by reference in their entireties. For example, the Fleximer® linker technology developed by Mersana et al. has the potential to enable high-DAR ADCs with good physicochemical properties. As shown below, the Fleximer® linker technology is based on incorporating drug molecules into a solubilizing poly-acetal backbone via a sequence of ester bonds The methodology renders highly-loaded ADCs (DAR up to 20) whilst maintaining good physicochemical properties. This methodology could be utilized with Bcl-xL inhibitors as shown in the Scheme below.
.1 v
Figure imgf000115_0001
4 4
92 99 24 E1 To utilize the Fleximer® linker technology depicted in the scheme above, an aliphatic alcohol can be present or introduced into the Bcl-xL inhibitor. The alcohol moiety is then conjugated to an alanine moiety, which is then synthetically incorporated into the Fleximer® linker. Liposomal processing of the ADC in vitro releases the parent alcohol–containing drug.
Additional examples of dendritic type linkers can be found in US 2006/116422; US
2005/271615; de Groot et al., (2003) Angew. Chem. Int. Ed.42:4490-4494; Amir et al., (2003) Angew. Chem. Int. Ed.42:4494-4499; Shamis et al., (2004) J. Am. Chem. Soc.126:1726-1731 ; Sun et al., (2002) Bioorganic & Medicinal Chemistry Letters 12:2213-2215; Sun et al., (2003) Bioorganic & Medicinal Chemistry 11:1761-1768; King et al., (2002) Tetrahedron Letters 43:1987-1990.
Exemplary monovalent linkers that may be used are described, for example, in Nolting, 2013, Antibody-Drug Conjugates, Methods in Molecular Biology 1045:71-100; Kitson et al., 2013, CROs/CMOs - Chemica Oggi– Chemistry Today 31(4): 30-36; Ducry et al., 2010, Bioconjugate Chem.21:5-13; Zhao et al., 2011, J. Med. Chem.54:3606-3623; U.S. Patent No.7,223,837; U.S. Patent No.8,568,728; U.S. Patent No.8,535,678; and WO2004010957, the content of each of which is incorporated herein by reference in their entireties.
By way of example and not limitation, some cleavable and noncleavable linkers that may be included in the ADCs described herein are described below.
Cleavable Linkers
In certain embodiments, the linker selected is cleavable in vitro and in vivo. Cleavable linkers may include chemically or enzymatically unstable or degradable linkages. Cleavable linkers generally rely on processes inside the cell to liberate the drug, such as reduction in the cytoplasm, exposure to acidic conditions in the lysosome, or cleavage by specific proteases or other enzymes within the cell. Cleavable linkers generally incorporate one or more chemical bonds that are either chemically or enzymatically cleavable while the remainder of the linker is noncleavable.
In certain embodiments, a linker comprises a chemically labile group such as hydrazone and/or disulfide groups. Linkers comprising chemically labile groups exploit differential properties between the plasma and some cytoplasmic compartments. The intracellular conditions to facilitate drug release for hydrazone containing linkers are the acidic environment of endosomes and lysosomes, while the disulfide containing linkers are reduced in the cytosol, which contains high thiol concentrations, e.g., glutathione. In certain embodiments, the plasma stability of a linker comprising a chemically labile group may be increased by introducing steric hindrance using substituents near the chemically labile group.
Acid-labile groups, such as hydrazone, remain intact during systemic circulation in the blood’s neutral pH environment (pH 7.3-7.5) and undergo hydrolysis and release the drug once the ADC is internalized into mildly acidic endosomal (pH 5.0-6.5) and lysosomal (pH 4.5-5.0) compartments of the cell. This pH dependent release mechanism has been associated with nonspecific release of the drug To increase the stability of the hydrazone group of the linker the linker may be varied by chemical modification, e.g., substitution, allowing tuning to achieve more efficient release in the lysosome with a minimized loss in circulation.
Hydrazone-containing linkers may contain additional cleavage sites, such as additional acid- labile cleavage sites and/or enzymatically labile cleavage sites. ADCs including exemplary hydrazone-containing linkers include the following structures:
Figure imgf000117_0001
Figure imgf000117_0002
Figure imgf000117_0003
wherein D and Ab represent the drug and Ab, respectively, and n represents the number of drug- linkers linked to the anti-B7-H3 antibody. In certain linkers such as linker (Ig), the linker comprises two cleavable groups– a disulfide and a hydrazone moiety. For such linkers, effective release of the unmodified free drug requires acidic pH or disulfide reduction and acidic pH. Linkers such as (Ih) and (Ii) have been shown to be effective with a single hydrazone cleavage site.
Other acid-labile groups that may be included in linkers include cis-aconityl-containing linkers. cis-Aconityl chemistry uses a carboxylic acid juxtaposed to an amide bond to accelerate amide hydrolysis under acidic conditions.
Cleavable linkers may also include a disulfide group. Disulfides are thermodynamically stable at physiological pH and are designed to release the drug upon internalization inside cells, wherein the cytosol provides a significantly more reducing environment compared to the extracellular environment. Scission of disulfide bonds generally requires the presence of a cytoplasmic thiol cofactor, such as (reduced) glutathione (GSH), such that disulfide-containing linkers are reasonable stable in circulation, selectively releasing the drug in the cytosol. The intracellular enzyme protein disulfide isomerase, or similar enzymes capable of cleaving disulfide bonds, may also contribute to the preferential cleavage of disulfide bonds inside cells. GSH is reported to be present in cells in the concentration range of 0.5-10 mM compared with a significantly lower concentration of GSH or cysteine, the most abundant low-molecular weight thiol, in circulation at approximately 5 µM. Tumor cells, where irregular blood flow leads to a hypoxic state, result in enhanced activity of reductive enzymes and therefore even higher glutathione concentrations. In certain embodiments, the in vivo stability of a disulfide-containing linker may be enhanced by chemical modification of the linker, e.g., use of steric hindrance adjacent to the disulfide bond.
ADCs including exemplary disulfide-containing linkers include the following structures:
Figure imgf000118_0001
Figure imgf000118_0002
(Il)
Figure imgf000118_0003
wherein D and Ab represent the drug and antibody, respectively, n represents the number of drug- linkers linked to the anti-B7-H3 antibody and R is independently selected at each occurrence from hydrogen or alkyl, for example. In certain embodiments, increasing steric hindrance adjacent to the disulfide bond increases the stability of the linker. Structures such as (Ij) and (Il) show increased in vivo stability when one or more R groups is selected from a lower alkyl such as methyl.
Another type of linker that may be used is a linker that is specifically cleaved by an enzyme. In one embodiment, the linker is cleavable by a lysosomal enzyme. Such linkers are typically peptide- based or include peptidic regions that act as substrates for enzymes. Peptide based linkers tend to be more stable in plasma and extracellular milieu than chemically labile linkers. Peptide bonds generally have good serum stability, as lysosomal proteolytic enzymes have very low activity in blood due to endogenous inhibitors and the unfavorably high pH value of blood compared to lysosomes. Release of a drug from an anti-B7-H3 antibody occurs specifically due to the action of lysosomal proteases, e.g., cathepsin and plasmin. These proteases may be present at elevated levels in certain tumor tissues. In certain embodiments, the linker is cleavable by a lysosomal enzyme. In certain embodiments, the linker is cleavable by a lysosomal enzyme, and the lysosomal enzyme is Cathepsin B. In certain embodiments, the linker is cleavable by a lysosomal enzyme, and the lysosomal enzyme is β-glucuronidase or β-galactosidase. In certain embodiments, the linker is cleavable by a lysosomal enzyme, and the lysosomal enzyme is β-glucuronidase. In certain embodiments, the linker is cleavable by a lysosomal enzyme, and the lysosomal enzyme is β-galactosidase.
In exemplary embodiments, the cleavable peptide is selected from tetrapeptides such as Gly-Phe-Leu-Gly, Ala-Leu-Ala-Leu or dipeptides such as Val-Cit, Val-Ala, and Phe-Lys. In certain embodiments, dipeptides are preferred over longer polypeptides due to hydrophobicity of the longer peptides.
A variety of dipeptide-based cleavable linkers useful for linking drugs such as doxorubicin, mitomycin, camptothecin, tallysomycin and auristatin/auristatin family members to antibodies have been described (see, Dubowchik et al., 1998, J. Org. Chem.67:1866-1872; Dubowchik et al., 1998, Bioorg. Med. Chem. Lett.8:3341-3346; Walker et al., 2002, Bioorg. Med. Chem. Lett.12:217-219; Walker et al., 2004, Bioorg. Med. Chem. Lett.14:4323-4327; and Francisco et al., 2003, Blood 102:1458-1465, the contents of each of which are incorporated herein by reference). All of these dipeptide linkers, or modified versions of these dipeptide linkers, may be used in the ADCs described herein. . Other dipeptide linkers that may be used include those found in ADCs such as Seattle Genetics’ Brentuximab Vendotin SGN-35 (Adcetris™), Seattle Genetics SGN-75 (anti-CD-70, MC- monomethyl auristatin F(MMAF), Celldex Therapeutics glembatumumab (CDX-011) (anti-NMB, Val-Cit- monomethyl auristatin E(MMAE), and Cytogen PSMA-ADC (PSMA-ADC-1301) (anti- PSMA, Val-Cit-MMAE).
Enzymatically cleavable linkers may include a self-immolative spacer to spatially separate the drug from the site of enzymatic cleavage. The direct attachment of a drug to a peptide linker can result in proteolytic release of an amino acid adduct of the drug, thereby impairing its activity. The use of a self-immolative spacer allows for the elimination of the fully active, chemically unmodified drug upon amide bond hydrolysis.
One self-immolative spacer is the bifunctional para-aminobenzyl alcohol group, which is linked to the peptide through the amino group, forming an amide bond, while amine containing drugs may be attached through carbamate functionalities to the benzylic hydroxyl group of the linker (to give a p-amidobenzylcarbamate, PABC). The resulting prodrugs are activated upon protease-mediated cleavage, leading to a 1,6-elimination reaction releasing the unmodified drug, carbon dioxide, and remnants of the linker group. The following scheme depicts the fragmentation of p-amidobenzyl carbamate and release of the drug:
Figure imgf000119_0001
wherein X-D represents the unmodified drug. Heterocyclic variants of this self-immolative group have also been described. See U.S. Patent No. 7,989,434.
In certain embodiments, the enzymatically cleavable linker is a ß-glucuronic acid-based linker. Facile release of the drug may be realized through cleavage of the ß-glucuronide glycosidic bond by the lysosomal enzyme ß-glucuronidase. This enzyme is present abundantly within lysosomes and is overexpressed in some tumor types, while the enzyme activity outside cells is low. ß- Glucuronic acid-based linkers may be used to circumvent the tendency of an ADC to undergo aggregation due to the hydrophilic nature of ß-glucuronides. In certain embodiments, ß-glucuronic acid-based linkers are preferred as linkers for ADCs linked to hydrophobic drugs. The following scheme depicts the release of the drug from and ADC containing a ß-glucuronic acid-based linker:
Figure imgf000120_0001
A variety of cleavable ß-glucuronic acid-based linkers useful for linking drugs such as auristatins, camptothecin and doxorubicin analogues, CBI minor-groove binders, and psymberin to antibodies have been described (see, Jeffrey et al., 2006, Bioconjug. Chem.17:831-840; Jeffrey et al., 2007, Bioorg. Med. Chem. Lett.17:2278-2280; and Jiang et al., 2005, J. Am. Chem. Soc.127:11254- 11255, the contents of each of which are incorporated herein by reference). All of these ß-glucuronic acid-based linkers may be used in the ADCs described herein. In certain embodiments, the enzymatically cleavable linker is a ß-galactoside-based linker. ß-Galactoside is present abundantly within lysosomes, while the enzyme activity outside cells is low.
Additionally, Bc1-xL inhibitors containing a phenol group can be covalently bonded to a linker through the phenolic oxygen. One such linker, described in U.S. Patent App. No.
2009/0318668, relies on a methodology in which a diamino-ethane“SpaceLink” is used in conjunction with traditional“PABO”-based self-immolative groups to deliver phenols. The cleavage of the linker is depicted schematically below using a Bcl-xL inhibitor of the disclosure.
Figure imgf000121_0001
.1 v 44 92 99 24 E1 Cleavable linkers may include noncleavable portions or segments, and/or cleavable segments or portions may be included in an otherwise non-cleavable linker to render it cleavable. By way of example only, polyethylene glycol (PEG) and related polymers may include cleavable groups in the polymer backbone. For example, a polyethylene glycol or polymer linker may include one or more cleavable groups such as a disulfide, a hydrazone or a dipeptide.
Other degradable linkages that may be included in linkers include ester linkages formed by the reaction of PEG carboxylic acids or activated PEG carboxylic acids with alcohol groups on a biologically active agent, wherein such ester groups generally hydrolyze under physiological conditions to release the biologically active agent. Hydrolytically degradable linkages include, but are not limited to, carbonate linkages; imine linkages resulting from reaction of an amine and an aldehyde; phosphate ester linkages formed by reacting an alcohol with a phosphate group; acetal linkages that are the reaction product of an aldehyde and an alcohol; orthoester linkages that are the reaction product of a formate and an alcohol; and oligonucleotide linkages formed by a
phosphoramidite group, including but not limited to, at the end of a polymer, and a 5' hydroxyl group of an oligonucleotide.
In certain embodiments, the linker comprises an enzymatically cleavable peptide moiety, for example, a linker comprising structural formula (IVa), (IVb), (IVc), or (IVd):
Figure imgf000122_0001
(IVd)
Figure imgf000123_0001
or a pharmaceutically acceptable salt thereof, wherein:
peptide represents a peptide (illustrated N→C, wherein peptide includes the amino and carboxy“termini”) a cleavable by a lysosomal enzyme;
T represents a polymer comprising one or more ethylene glycol units or an alkylene chain, or combinations thereof;
Ra is selected from hydrogen, C1-6 alkyl, SO3H and CH2SO3H;
Ry is hydrogen or C1-4 alkyl-(O)r-(C1-4 alkylene)s-G1 or C1-4 alkyl-(N)-[(C1-4 alkylene)-G1]2; Rz is C1-4 alkyl-(O)r-(C1-4 alkylene)s-G2;
G1 is SO3H, CO2H, PEG 4-32, or sugar moiety;
G2 is SO3H, CO2H, or PEG 4-32 moiety;
r is 0 or 1;
s is 0 or 1;
p is an integer ranging from 0 to 5;
q is 0 or 1;
x is 0 or 1;
y is 0 or 1;
represents the point of attachment of the linker to the Bcl-xL inhibitor; and
* represents the point of attachment to the remainder of the linker.
In certain embodiments, the linker comprises an enzymatically cleavable peptide moiety, for example, a linker comprising structural formula (IVa), (IVb), (IVc), (IVd) or a pharmaceutically acceptable salt thereof.
In certain embodiments, linker L comprises a segment according to structural formula IVa or IVb or a pharmaceutically acceptable salt thereof.
In certain embodiments, the peptide is selected from a tripeptide or a dipeptide. In particular embodiments, the dipeptide is selected from: Val-Cit; Cit-Val; Ala-Ala; Ala-Cit; Cit-Ala; Asn-Cit; Cit-Asn; Cit-Cit; Val-Glu; Glu-Val; Ser-Cit; Cit-Ser; Lys-Cit; Cit-Lys; Asp-Cit; Cit-Asp; Ala-Val; Val-Ala; Phe-Lys; Lys-Phe; Val-Lys; Lys-Val; Ala-Lys; Lys-Ala; Phe-Cit; Cit-Phe; Leu-Cit; Cit-Leu; Ile-Cit; Cit-Ile; Phe-Arg; Arg-Phe; Cit-Trp; and Trp-Cit, or a pharmaceutically acceptable salt thereof.
Exemplary embodiments of linkers according to structural formula (IVa) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an antibody):
Figure imgf000124_0001
Figure imgf000124_0002
Figure imgf000124_0003
Figure imgf000124_0004
Figure imgf000124_0005
(IVa.6)
Figure imgf000124_0006
Figure imgf000125_0001
Figure imgf000125_0002
Exemplary embodiments of linkers according to structural formula (IVb), (IVc), or (IVd) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an antibody):
Figure imgf000125_0003
Figure imgf000125_0004
Figure imgf000125_0005
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000127_0002
Figure imgf000127_0003
Figure imgf000127_0004
Figure imgf000127_0005
Figure imgf000128_0001
Figure imgf000128_0002
Figure imgf000128_0003
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000130_0002
(IVd.1)
Figure imgf000130_0003
Figure imgf000130_0004
Figure imgf000131_0001
Figure imgf000131_0002
In certain embodiments, the linker comprises an enzymatically cleavable sugar moiety, for example, a linker comprising structural formula (Va), (Vb), (Vc), (Vd), or (Ve)::
Figure imgf000131_0003
Figure imgf000131_0004
Figure imgf000132_0001
Figure imgf000132_0002
Figure imgf000132_0003
or a pharmaceuticall acceptable salt thereof, wherein:
q is 0 or 1;
r is 0 or 1;
X1 is CH2, O or NH;
represents the point of attachment of the linker to the drug; and * represents the point of attachment to the remainder of the linker.
Exemplary embodiments of linkers according to structural formula (Va) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an anti-B7-H3 antibody):
Figure imgf000133_0001
Figure imgf000133_0002
Figure imgf000133_0003
Figure imgf000133_0004
(Va.5)
Figure imgf000133_0005
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000135_0002
Exemplary embodiments of linkers according to structural formula (Vb) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an anti-B7-H3 antibody):
Figure imgf000135_0003
Figure imgf000135_0004
Figure imgf000136_0001
Figure imgf000136_0002
Figure imgf000136_0003
Figure imgf000136_0004
(Vb.7)
Figure imgf000137_0001
Figure imgf000137_0002
Figure imgf000137_0003
Figure imgf000137_0004
Exemplary embodiments of linkers according to structural formula (Vc) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an anti-B7-H3 antibody):
Figure imgf000138_0001
Figure imgf000138_0002
Figure imgf000138_0003
(Vc.4)
Figure imgf000138_0004
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000140_0002
Figure imgf000140_0003
Exemplary embodiments of linkers according to structural formula (Vd) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an anti-B7-H3 antibody):
Figure imgf000140_0004
Figure imgf000141_0001
(Vd.5)
Figure imgf000141_0002
Figure imgf000142_0001
Exemplary embodiments of linkers according to structural formula (Ve) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an anti-B7-H3 antibody):
Figure imgf000142_0002
Non-Cleavable Linkers
Although cleavable linkers may provide certain advantages, the linkers comprising the ADC described herein need not be cleavable. For noncleavable linkers, the drug release does not depend on the differential properties between the plasma and some cytoplasmic compartments. The release of the drug is postulated to occur after internalization of the ADC via antigen-mediated endocytosis and delivery to lysosomal compartment, where the anti-B7-H3 antibody is degraded to the level of amino acids through intracellular proteolytic degradation. This process releases a drug derivative, which is formed by the drug, the linker, and the amino acid residue to which the linker was covalently attached. The amino-acid drug metabolites from conjugates with noncleavable linkers are more hydrophilic and generally less membrane permeable, which leads to less bystander effects and less nonspecific toxicities compared to conjugates with a cleavable linker. In general, ADCs with noncleavable linkers have greater stability in circulation than ADCs with cleavable linkers. Non- cleavable linkers may be alkylene chains, or maybe polymeric in natures, such as, for example, based upon polyalkylene glycol polymers, amide polymers, or may include segments of alkylene chains, polyalkylene glycols and/or amide polymers. In certain embodiments, the linker comprises a polyethylene glycol segment having from 1 to 6 ethylene glycol units.
A variety of non-cleavable linkers used to link drugs to antibodies have been described. (See, Jeffrey et al., 2006, Bioconjug. Chem.17:831-840; Jeffrey et al., 2007, Bioorg. Med. Chem. Lett. 17:2278-2280; and Jiang et al., 2005, J. Am . Chem. Soc.127:11254-11255, the contents of which are incorporated herein by reference). All of these linkers may be included in the ADCs described herein.
In certain embodiments, the linker is non-cleavable in vivo, for example a linker according to structural formula (VIa), (VIb), (VIc) or (VId) (as illustrated, the linkers include a group suitable for covalently linking the linker to an anti-B7-H3 antibody:
Figure imgf000143_0001
Figure imgf000143_0002
Figure imgf000143_0003
Figure imgf000143_0004
or a pharmaceutically acceptable salt thereof, wherein:
Ra is selected from hydrogen, alkyl, sulfonate and methyl sulfonate; Rx is a moiety including a functional group capable of covalently linking the linker to an antibody; and
represents the point of attachment of the linker to the Bcl-xL inhibitor.
Exemplary embodiments of linkers according to structural formula (VIa)-(VId) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an anti-B7-H3 antibody, and“ “ represents the point of attachment to a Bcl-xL inhibitor):
Figure imgf000144_0001
Figure imgf000144_0002
Figure imgf000144_0003
Figure imgf000144_0004
Figure imgf000144_0005
Figure imgf000144_0006
Figure imgf000145_0001
Groups Used to Attach Linkers to Anti-B7-H3 Antibodies
Attachment groups can be electrophilic in nature and include: maleimide groups, activated disulfides, active esters such as NHS esters and HOBt esters, haloformates, acid halides, alkyl and benzyl halides such as haloacetamides. As discussed below, there are also emerging technologies related to“self-stabilizing” maleimides and“bridging disulfides” that can be used in accordance with the disclosure.
One example of a“self-stabilizing” maleimide group that hydrolyzes spontaneously under antibody conjugation conditions to give an ADC species with improved stability is depicted in the schematic below. See U.S. Published Application No.2013/0309256, International Application Publication No. WO 2013/173337, Tumey et al., 2014, Bioconjugate Chem.25: 1871-1880, and Lyon et al., 2014, Nat. Biotechnol.32: 1059-1062. Thus, the maleimide attachment group is reacted with a sulfhydryl of an antibody to give an intermediate succinimide ring. The hydrolyzed form of the attachment group is resistant to deconjugation in the presence of plasma proteins.
Figure imgf000146_0001
.1 v 44 92 99 24 E1 As shown above, the maleimide ring of a linker may react with an antibody Ab, forming a covalent attachment as either a succinimide (closed form) or succinamide (open form).
Polytherics has disclosed a method for bridging a pair of sulfhydryl groups derived from reduction of a native hinge disulfide bond. See, Badescu et al., 2014, Bioconjugate Chem.25:1124- 1136. The reaction is depicted in the schematic below. An advantage of this methodology is the ability to synthesize homogenous DAR4 ADCs by full reduction of IgGs (to give 4 pairs of sulfhydryls) followed by reaction with 4 equivalents of the alkylating agent. ADCs containing “bridged disulfides” are also claimed to have increased stability.
Figure imgf000148_0001
.1 v 44 92 99 24 E1 Similarly, as depicted below, a maleimide derivative that is capable of bridging a pair of sulfhydryl groups has been developed. See U.S. Published Application No.2013/0224228.
Figure imgf000149_0001
In certain embodiments the attachment moiety comprises the structural formulae (VIIa), (VIIb), or (VIIc):
Figure imgf000149_0002
or a pharmaceutically acceptable salt thereof, wherein:
Rq is H or–O-(CH2CH2O)11-CH3;
x is 0 or 1;
y is 0 or 1;
G3 is–CH2CH2CH2SO3H or–CH2CH2O-(CH2CH2O)11-CH3;
Rw is–O-CH2CH2SO3H or–NH(CO)-CH2CH2O-(CH2CH2O)12-CH3; and
* represents the point of attachment to the remainder of the linker. In certain embodiments, the linker comprises a segment according to structural formulae III III r III
(VIIIc)
Figure imgf000150_0001
or a hydrolyzed derivative or a pharmaceutically acceptable salt thereof, wherein:
Rq is H or–O-(CH2CH2O)11-CH3;
x is 0 or 1;
y is 0 or 1;
G3 is–CH2CH2CH2SO3H or–CH2CH2O-(CH2CH2O)11-CH3;
Rw is–O-CH2CH2SO3H or–NH(CO)-CH2CH2O-(CH2CH2O)12-CH3;
* represents the point of attachment to the remainder of the linker; and
represents the point of attachment of the linker to the antibody, wherein when in the hydrolyzed form, can be either at the α-position or β-position of the carboxylic acid next to it.
Exemplary embodiments of linkers according to structural formula (VIIa) and (VIIb) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an antibody):
Figure imgf000151_0001
.1 v 44 92 99 24 E1
Figure imgf000152_0001
.1 v 44 92 99 24 E1
Figure imgf000153_0001
.1 v 44 92 99 24 E1
Figure imgf000154_0001
.1 v 44 92 99 24 E1
Figure imgf000155_0001
.1 v 44 92 99 24 E1
Figure imgf000156_0001
.1 v 44 92 99 24 E1
Figure imgf000157_0001
.1 v 44 92 99 24 E1 Exemplary embodiments of linkers according to structural formula (VIIc) that may be included in the ADCs described herein include the linkers illustrated below (as illustrated, the linkers include a group suitable for covalently linking the linker to an antibody):
Figure imgf000158_0001
Figure imgf000158_0002
Figure imgf000159_0001
(VIIc.4)
Figure imgf000159_0002
(VIIc.5)
Figure imgf000159_0003
Figure imgf000160_0001
In certain embodiments, L is selected from the group consisting of IVa.1-IVa.8, IVb.1- IVb.19, IVc.1-IVc.7, IVd.1-IVd.4, Va.1-Va.12, Vb.1-Vb.10, Vc.1-Vc.11, Vd.1-Vd.6, Ve.1-Ve.2, VIa.1, VIc.1-V1c.2, VId.1-VId.4, VIIa.1-VIIa.4, VIIb.1-VIIb.8, VIIc.1-VIIc.6 in either the closed or open form.
In certain embodiments, L is selected from the group consisting of IVb.2, IVc.5, IVc.6, IVc.7, IVd.4, Vb.9, Vc.11, VIIa.1, VIIa.3, VIIc.1, VIIc.4, and VIIc.5, wherein the maleimide of each linker has reacted with the antibody Ab, forming a covalent attachment as either a succinimide (closed form) or succinamide (open form).
In certain embodiments, linker L is selected from the group consisting of IVb.2, IVc.5, IVc.6, IVd.4, Vc.11, VIIa.1, VIIa.3, VIIc.1, VIIc.4, VIIc.5, wherein the maleimide of each linker has reacted with the antibody Ab, forming a covalent attachment as either a succinimide (closed form) or succinamide (open form).
In certain embodiments, linker L is selected from the group consisting of IVb.2, Vc.11, VIIa.3, IVc.6, and VIIc.1, wherein is the attachment point to drug D and @ is the attachment point to the LK, wherein when the linker is in the open form as shown below, @ can be either at the α- position or β-position of the carboxylic acid next to it:
Figure imgf000160_0002
,
Figure imgf000161_0001
,
,
Figure imgf000162_0001
Figure imgf000163_0001
. Bcl-xL Linker Selection Considerations
As is known by skilled artisans, the linker selected for a particular ADC may be influenced by a variety of factors, including but not limited to, the site of attachment to the antibody (e.g., lys, cys or other amino acid residues), structural constraints of the drug pharmacophore and the lipophilicity of the drug. The specific linker selected for an ADC should seek to balance these different factors for the specific antibody/drug combination. For a review of the factors that are influenced by choice of linkers in ADCs, see Nolting, Chapter 5“Linker Technology in Antibody-Drug Conjugates,” In: Antibody-Drug Conjugates: Methods in Molecular Biology, vol.1045, pp.71-100, Laurent Ducry (Ed.), Springer Science & Business Medica, LLC, 2013.
For example, ADCs have been observed to effect killing of bystander antigen-negative cells present in the vicinity of the antigen-positive tumor cells. The mechanism of bystander cell killing by ADCs has indicated that metabolic products formed during intracellular processing of the ADCs may play a role. Neutral cytotoxic metabolites generated by metabolism of the ADCs in antigen-positive cells appear to play a role in bystander cell killing while charged metabolites may be prevented from diffusing across the membrane into the medium and therefore cannot affect bystander killing. In certain embodiments, the linker is selected to attenuate the bystander killing effect caused by cellular metabolites of the ADC. In certain embodiments, the linker is selected to increase the bystander killing effect.
The properties of the linker may also impact aggregation of the ADC under conditions of use and/or storage. Typically, ADCs reported in the literature contain no more than 3-4 drug molecules per antibody molecule (see, e.g., Chari, 2008, Acc Chem Res 41:98-107). Attempts to obtain higher drug-to-antibody ratios (“DAR”) often failed, particularly if both the drug and the linker were hydrophobic, due to aggregation of the ADC (King et al., 2002, J Med Chem 45:4336-4343;
Hollander et al., 2008, Bioconjugate Chem 19:358-361; Burke et al., 2009 Bioconjugate Chem 20:1242-1250). In many instances, DARs higher than 3-4 could be beneficial as a means of increasing potency. In instances where the Bcl-xL inhibitor is hydrophobic in nature, it may be desirable to select linkers that are relatively hydrophilic as a means of reducing ADC aggregation, especially in instances where DARS greater than 3-4 are desired. Thus, in certain embodiments, the linker incorporates chemical moieties that reduce aggregation of the ADCs during storage and/or use. A linker may incorporate polar or hydrophilic groups such as charged groups or groups that become charged under physiological pH to reduce the aggregation of the ADCs. For example, a linker may incorporate charged groups such as salts or groups that deprotonate, e.g., carboxylates, or protonate, e.g., amines, at physiological pH.
Exemplary polyvalent linkers that have been reported to yield DARs as high as 20 that may be used to link numerous Bcl-xL inhibitors to an antibody are described in U.S. Patent No 8,399,512; U.S. Published Application No.2010/0152725; U.S. Patent No.8,524,214; U.S. Patent No.8,349,308; U.S. Published Application No.2013/189218; U.S. Published Application No. 2014/017265; WO 2014/093379; WO 2014/093394; WO 2014/093640, the content of which are incorporated herein by reference in their entireties.
In particular embodiments, the aggregation of the ADCs during storage or use is less than about 40% as determined by size-exclusion chromatography (SEC). In particular embodiments, the aggregation of the ADCs during storage or use is less than 35%, such as less than about 30%, such as less than about 25%, such as less than about 20%, such as less than about 15%, such as less than about 10%, such as less than about 5%, such as less than about 4%, or even less, as determined by size- exclusion chromatography (SEC).
III.A.3. Bcl-xL ADC Synthons
Antibody-Drug Conjugate synthons are synthetic intermediates used to form ADCs. The synthons are generally compounds according to structural formula (III):
Figure imgf000164_0001
or salts thereof, wherein D is a Bcl-xL inhibitor as previously described, L is a linker as previously described, and Rx is a reactive group suitable for linking the synthon to an antibody. In specific embodiments, the ADC synthons are compounds according to structural formulae (IIIa) and (IIIb) , or salts thereof, where the various substituents are as previously defined for structural formulae (IIa) and (IIb), respectively, and L and Rx are as defined for structural formula (III):
Figure imgf000165_0001
Figure imgf000165_0002
To synthesize an ADC, an intermediate synthon according to structural formula (III), or a salt thereof, is contacted with an antibody of interest under conditions in which functional group Rx reacts with a“complementary” functional group on the antibody, Fx, to form a covalent linkage.
Figure imgf000165_0003
The identities of groups Rx and Fx will depend upon the chemistry used to link the synthon to the antibody. Generally, the chemistry used should not alter the integrity of the antibody, for example its ability to bind its target. Preferably, the binding properties of the conjugated antibody will closely resemble those of the unconjugated antibody. A variety of chemistries and techniques for conjugating molecules to biological molecules such as antibodies are known in the art and in particular to antibodies, are well-known. See, e.g., Amon et al.,“Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy,” in: Monoclonal Antibodies And Cancer Therapy, Reisfeld et al., Eds., Alan R. Liss, Inc., 1985; Hellstrom et al.,“Antibodies For Drug Delivery,” in: Controlled Drug Delivery, Robinson et al., Eds., Marcel Dekker, Inc., 2nd Ed.1987; Thorpe,“Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review,” in: Monoclonal Antibodies '84: Biological And Clinical Applications, Pinchera et al., Eds., 1985;“Analysis, Results, and Future Prospective of the Therapeutic Use of Radiolabeled Antibody In Cancer Therapy,” in: Monoclonal Antibodies For Cancer Detection And Therapy, Baldwin et al., Eds., Academic Press, 1985; Thorpe et al., 1982, Immunol. Rev.62:119-58; PCT publication WO 89/12624. Any of these chemistries may be used to link the synthons to an antibody. In one embodiment, Rx comprises a functional group capable of linking the synthon to an amino group on an antibody. In another embodiment, Rx comprises an NHS-ester or an
isothiocyanate. In another embodiment, Rx comprises a functional group capable of linking the synthon to a sulfhydryl group on an antibody. In another embodiment, Rx comprises a haloacetyl or a maleimide. In another embodiment, L is selected from IVa or IVb and salts thereof; and Rx comprises a functional group selected from the group consisting of NHS-ester, isothiocyanate, haloacetyl and maleimide.
Typically, the synthons are linked to the side chains of amino acid residues of the antibody, including, for example, the primary amino group of accessible lysine residues or the sulfhydryl group of accessible cysteine residues. Free sulfhydryl groups may be obtained by reducing interchain disulfide bonds.
In one embodiment, LK is a linkage formed with an amino group on the anti-B7-H3 antibody Ab (e.g., huAb13v1, huAb3v2.5, or huAb3v2.6). In another embodiment, LK is an amide or a thiourea. In another embodiment, LK is a linkage formed with a sulfhydryl group on the anti-B7-H3 antibody Ab. In another embodiment, LK is a thioether.
In one embodiment, LK is selected from the group consisting of amide, thiourea and thioether; and m is an integer ranging from 1 to 8.
A number of functional groups Rx and chemistries useful for linking synthons to accessible lysine residues are known, and include by way of example and not limitation NHS-esters and isothiocyanates.
A number of functional groups Rx and chemistries useful for linking synthons to accessible free sulfhydryl groups of cysteine residues are known, and include by way of example and not limitation haloacetyls and maleimides.
However, conjugation chemistries are not limited to available side chain groups. Side chains such as amines may be converted to other useful groups, such as hydroxyls, by linking an appropriate small molecule to the amine. This strategy can be used to increase the number of available linking sites on the antibody by conjugating multifunctional small molecules to side chains of accessible amino acid residues of the antibody. Functional groups Rx suitable for covalently linking the synthons to these“converted” functional groups are then included in the synthons.
The antibody may also be engineered to include amino acid residues for conjugation. An approach for engineering antibodies to include non-genetically encoded amino acid residues useful for conjugating drugs in the context of ADCs is described in Axup et al., 2003, Proc Natl Acad Sci 109:16101-16106 and Tian et al., 2014, Proc Natl Acad Sci 111:1776-1771, as are chemistries and functional group useful for linking synthons to the non-encoded amino acids.
Exemplary synthons useful for making ADCs described herein include, but are not limited to, the following synthons listed below in Table B.
.1 v 44 92 99 24 E1
Figure imgf000167_0001
.1 v 44 92 99 24 E1
Figure imgf000168_0001
.1 v 44 92 99 24 E1
Figure imgf000169_0001
.1 v 44 92 99 24 E1
Figure imgf000170_0001
.1 v 44 92 99 24 E1
Figure imgf000171_0001
.1 v 44 92 99 24 E1
Figure imgf000172_0001
.1 v 44 92 99 24 E1
Figure imgf000173_0001
.1 v 44 92 99 24 E1
Figure imgf000174_0001
.1 v 44 92 99 24 E1
Figure imgf000175_0001
.1 v 44 92 99 24 E1
Figure imgf000176_0001
.1 v 44 92 99 24 E1
Figure imgf000177_0001
.1 v 44 92 99 24 E1
Figure imgf000178_0001
.1 v 44 92 99 24 E1
.1 v 44 92 99 24 E1
Figure imgf000180_0001
.1 v 44 92 99 24 E1
Figure imgf000181_0001
.1 v 44 92 99 24 E1
Figure imgf000182_0001
.1 v 44 92 99 24 E1
Figure imgf000183_0001
.1 v 44 92 99 24 E1
Figure imgf000184_0001
.1 v 44 92 99 24 E1
Figure imgf000185_0001
.1 v 44 92 99 24 E1
Figure imgf000186_0001
.1 v 44 92 99 24 E1
Figure imgf000187_0001
.1 v 44 92 99 24 E1
Figure imgf000188_0001
.1 v 44 92 99 24 E1
Figure imgf000189_0001
.1 v 44 92 99 24 E1
Figure imgf000190_0001
.1 v 44 92 99 24 E1
Figure imgf000191_0001
.1 v 44 92 99 24 E1
Figure imgf000192_0001
.1 v 44 92 99 24 E1
Figure imgf000193_0001
.1 v 44 92 99 24 E1
Figure imgf000194_0001
.1 v 44 92 99 24 E1
Figure imgf000195_0001
.1 v 44 92 99 24 E1
Figure imgf000196_0001
.1 v 44 92 99 24 E1
Figure imgf000197_0001
.1 v 44 92 99 24 E1
Figure imgf000198_0001
.1 v 44 92 99 24 E1
Figure imgf000199_0001
.1 v 44 92 99 24 E1
Figure imgf000200_0001
.1 v 44 92 99 24 E1
Figure imgf000201_0001
.1 v 44 92 99 24 E1
Figure imgf000202_0001
.1 v 44 92 99 24 E1
Figure imgf000203_0001
1 v. 44 92 99 24 E1
Figure imgf000204_0001
.1 v 44 92 99 24 E1
Figure imgf000205_0001
1 v. 44 92 99 24 E1
Figure imgf000206_0001
In certain embodiments, the synthon is selected from the group consisting of synthon examples 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 2.10, 2.11, 2.12, 2.13, 2.14, 2.15, 2.16, 2.17, 2.18, 2.19, 2.20, 2.21, 2.22, 2.23, 2.24, 2.25, 2.26, 2.27, 2.28, 2.29, 2.30, 2.31, 2.34, 2.35, 2.36, 2.37, 2.38, 2.39, 2.40, 2.41, 2.42, 2.43, 2.44, 2.45, 2.46, 2.47, 2.48, 2.49, 2.50, 2.51, 2.52, 2.53, 2.54, 2.55, 2.56, 2.57, 2.58, 2.59, 2.60, 2.61, 2.62, 2.63, 2.64, 2.65, 2.66, 2.67, 2.68, 2.69, 2.70, 2.71, 2.72, and pharmaceutically acceptable salts thereof. The corresponding compound names of these synthons are provided below:
N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-L-valyl-N-{4-[({[2-({3-[(4- {6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4-tetrahydroquinolin-7-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]phenyl}-N5-carbamoyl-L-ornithinamide;
N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-L-valyl-N-{4-[({[2-({3-[(4- {6-[4-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-2H-1,4-benzoxazin-6-yl]-2-carboxypyridin-3- yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]phenyl}-N5-carbamoyl-L-ornithinamide;
N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-L-valyl-N-{4-[({[2-({3-[(4- {6-[4-(1,3-benzothiazol-2-ylcarbamoyl)-1-methyl-1,2,3,4-tetrahydroquinoxalin-6-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]phenyl}-N5-carbamoyl-L-ornithinamide;
4-[(1E)-3-({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4- tetrahydroquinolin-7-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](methyl)carbamoyl}oxy)prop-1-en-1-yl]-2-({N-[6- (2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-beta-alanyl}amino)phenyl beta-D- glucopyranosiduronic acid;
4-[(1E)-3-({[2-({3-[(4-{6-[4-(1,3-benzothiazol-2-ylcarbamoyl)-1-methyl-1,2,3,4- tetrahydroquinoxalin-6-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](methyl)carbamoyl}oxy)prop-1-en-1-yl]-2-({N-[6- (2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-beta-alanyl}amino)phenyl beta-D- glucopyranosiduronic acid;
4-[(1E)-3-({[2-({3-[(4-{6-[4-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-2H-1,4- benzoxazin-6-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](methyl)carbamoyl}oxy)prop-1-en-1-yl]-2-({N-[6- (2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-beta-alanyl}amino)phenyl beta-D- glucopyranosiduronic acid;
4-[(1E)-3-({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)prop-1-en-1-yl]-2-({N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl]-beta-alanyl}amino)phenyl beta-D-glucopyranosiduronic acid;
4-[(1E)-3-({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.1~3,7~]dec-1-yl}oxy)ethyl](oxetan-3-yl)carbamoyl}oxy)prop-1-en-1-yl]-2- ({N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-beta-alanyl}amino)phenyl beta-D- glucopyranosiduronic acid;
4-[(1E)-3-({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)prop-1-en-1-yl]-2- ({N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-beta-alanyl}amino)phenyl beta-D- glucopyranosiduronic acid;
4-[(1E)-3-({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)prop-1-en-1-yl]-2- ({N-[(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetyl]-beta-alanyl}amino)phenyl beta-D- glucopyranosiduronic acid;
4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-3-[2-(2-{[3- (2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)propanoyl]amino}ethoxy)ethoxy]phenyl beta-D- glucopyranosiduronic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]- 3-(1-{[3-(2-{[({(2E)-3-[4-{[(2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl]oxy}-3-({3-[({[(2E)-3-(4-{[(2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl]oxy}-3-[(3-{[3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)propanoyl]amino}propanoyl)amino]phenyl)prop-2-en-1- yl]oxy}carbonyl)amino]propanoyl}amino)phenyl]prop-2-en-1-yl}oxy)carbonyl](2- methoxyethyl)amino}ethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H-pyrazol- 4-yl)pyridine-2-carboxylic;
4-[({[2-(2-{2-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-5-(beta-D- glucopyranuronosyloxy)phenoxy}ethoxy)ethyl]carbamoyl}oxy)methyl]-3-[2-(2-{[3-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)propanoyl]amino}ethoxy)ethoxy]phenyl beta-D-glucopyranosiduronic acid;
4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-57- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-3-[2-(2- {[(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetyl]amino}ethoxy)ethoxy]phenyl beta-D- glucopyranosiduronic acid;
6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4-tetrahydroquinolin-7-yl]-3-{1-[(3- {[34-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)-3-methyl-4,32-dioxo-7,10,13,16,19,22,25,28-octaoxa- 3,31-diazatetratriacont-1-yl]oxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H- pyrazol-4-yl}pyridine-2-carboxylic acid;
4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-cyano-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl]carbamoyl}oxy)methyl]-3-[2-(2-{[3-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)propanoyl]amino}ethoxy)ethoxy]phenyl beta-D-glucopyranosiduronic acid;
4-[(1E)-3-({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)prop-1-en-1-yl]-2- ({N-[3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)propanoyl]-beta-alanyl}amino)phenyl beta-D- glucopyranosiduronic acid;
N-[(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetyl]-3-sulfo-L-alanyl-N-{5-[(1E)-3- ({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)prop-1-en-1-yl]-2-(beta-D- glucopyranuronosyloxy)phenyl}-beta-alaninamide;
N-[3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)propanoyl]-3-sulfo-L-alanyl-N-{5-[(1E)- 3-({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]- 2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)prop-1-en-1-yl]-2-(beta-D- glucopyranuronosyloxy)phenyl}-beta-alaninamide;
N-[(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetyl]-beta-alanyl-N-{5-[(1E)-3-({[2-({3- [(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)prop-1-en-1-yl]-2-(beta-D- glucopyranuronosyloxy)phenyl}-beta-alaninamide;
N-[3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)propanoyl]-beta-alanyl-N-{5-[(1E)-3- ({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)prop-1-en-1-yl]-2-(beta-D- glucopyranuronosyloxy)phenyl}-beta-alaninamide; 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-3-{2-[2-({N- [(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetyl]-3-sulfo-L-alanyl}amino)ethoxy]ethoxy}phenyl beta- D-glucopyranosiduronic acid;
4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-3-{2-[2-({N- [3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)propanoyl]-3-sulfo-L-alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid;
4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-3-{2-[2-({N- [(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetyl]-beta-alanyl}amino)ethoxy]ethoxy}phenyl beta-D- glucopyranosiduronic acid;
4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-3-{2-[2-({N- [3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)propanoyl]-beta-alanyl}amino)ethoxy]ethoxy}phenyl beta- D-glucopyranosiduronic acid;
2-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-5-{2-[2-({N- [3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)propanoyl]-3-sulfo-L-alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid;
2-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-5-{2-[2-({N- [6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L-alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid;
4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-3-[3-({N-[6- (2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L-alanyl}amino)propoxy]phenyl beta-D- glucopyranosiduronic acid;
4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-57- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-[3-({N-[6-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L-alanyl}amino)propoxy]phenyl beta-D- glucopyranosiduronic acid;
N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-L-valyl-N-{4-[({[(3S)-1-{8- (1,3-benzothiazol-2-ylcarbamoyl)-2-[6-carboxy-5-(1-{[3-(2-methoxyethoxy)-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl]-1,2,3,4- tetrahydroisoquinolin-6-yl}pyrrolidin-3-yl]carbamoyl}oxy)methyl]phenyl}-L-alaninamide;
N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-L-valyl-N-{4-[({[2-({3-[(4- {6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2- sulfamoylethyl)carbamoyl}oxy)methyl]phenyl}-N5-carbamoyl-L-ornithinamide;
4-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4-tetrahydroquinolin-7- yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-3-{2-[2-({N-[6-(2,5-dioxo-2,5-dihydro-1H- pyrrol-1-yl)hexanoyl]-3-sulfo-L-alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid;
2-[({[2-({3-[(4-{6-[5-(1,3-benzothiazol-2-ylcarbamoyl)quinolin-3-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-5-{2-[2-({N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl]-3-sulfo-L-alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid;
2-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4-tetrahydroquinolin-7- yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-5-[2-(2-{[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl]amino}ethoxy)ethoxy]phenyl beta-D-glucopyranosiduronic acid;
4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-{2-[2-({N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl]-3-sulfo-L-alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid;
2-[({[2-({3-[(4-{6-[4-(1,3-benzothiazol-2-ylcarbamoyl)quinolin-6-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-5-[2-(2-{[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl]amino}ethoxy)ethoxy]phenyl beta-D-glucopyranosiduronic acid;
4-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4-tetrahydroquinolin-7- yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-{2-[2-({N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl]-3-sulfo-L-alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid; 4-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4-tetrahydroquinolin-7- yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-(3-{[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl]amino}propoxy)phenyl beta-D-glucopyranosiduronic acid;
4-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4-tetrahydroquinolin-7- yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-[3-({N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl]-3-sulfo-L-alanyl}amino)propoxy]phenyl beta-D-glucopyranosiduronic acid;
2-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4-tetrahydroquinolin-7- yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-5-{2-[2-({N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl]-3-sulfo-L-alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid;
4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-3-{2-[2-({N-[6-(2,5-dioxo-2,5-dihydro-1H- pyrrol-1-yl)hexanoyl]-3-sulfo-L-alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid;
N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-L-valyl-N-[4-({[{2-[{8-(1,3- benzothiazol-2-ylcarbamoyl)-2-[6-carboxy-5-(1-{[3-(2-methoxyethoxy)-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl]-1,2,3,4- tetrahydroisoquinolin-6-yl}(methyl)amino]ethyl}(methyl)carbamoyl]oxy}methyl)phenyl]-L- alaninamide;
N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-L-valyl-N-{4-[({[2-({3-[(4- {6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-6-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]phenyl}-L-alaninamide;
2-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-5-{2-[2-({N-[6-(2,5-dioxo-2,5-dihydro-1H- pyrrol-1-yl)hexanoyl]-3-sulfo-L-alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid;
2-[({[2-({3-[(4-{6-[5-(1,3-benzothiazol-2-ylcarbamoyl)quinolin-3-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-5-[2-(2-{[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl]amino}ethoxy)ethoxy]phenyl beta-D-glucopyranosiduronic acid;
4-[({[2-({3-[(4-{6-[5-(1,3-benzothiazol-2-ylcarbamoyl)quinolin-3-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-57-dimethyltricyclo[33113,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-[2-(2-{[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl]amino}ethoxy)ethoxy]phenyl beta-D-glucopyranosiduronic acid;
6-[5-(1,3-benzothiazol-2-ylcarbamoyl)quinolin-3-yl]-3-(1-{[3-(2-{[6-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)hexanoyl](methyl)amino}ethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid;
4-[({[2-({3-[(4-{6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-1H-indol-2-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-2-({N-[3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)propanoyl]-beta-alanyl}amino)phenyl beta-D-glucopyranosiduronic acid;
4-[({[2-({3-[(4-{6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-1H-indol-2-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-[2-(2-{[3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)propanoyl]amino}ethoxy)ethoxy]phenyl beta-D-glucopyranosiduronic acid;
4-[({[2-({3-[(4-{6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-1H-indol-2-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-{2-[2-({N-[3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)propanoyl]-3-sulfo-L-alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid;
4-[({[2-({3-[(4-{6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-3-methyl-1H-indol-2-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-[2-(2-{[3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)propanoyl]amino}ethoxy)ethoxy]phenyl beta-D-glucopyranosiduronic acid;
N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-L-valyl-N-{4-[({[2-({3-[(4- {6-[4-(1,3-benzothiazol-2-ylcarbamoyl)isoquinolin-6-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]phenyl}-N5-carbamoyl-L-ornithinamide;
4-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-5,6-dihydroimidazo[1,5- a]pyrazin-7(8H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl]carbamoyl}oxy)methyl]-3-[2-(2-{[(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)acetyl]amino}ethoxy)ethoxy]phenyl beta-D-glucopyranosiduronic acid;
2-[({[2-({3-[(4-{6-[5-(1,3-benzothiazol-2-ylcarbamoyl)quinolin-3-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl]carbamoyl}oxy)methyl]-4-[19-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)-14-oxo-4,7,10- trioxa-13-azanonadec-1-yl]phenyl beta-D-glucopyranosiduronic acid;
4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-[4-({N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl]-3-sulfo-L-alanyl}amino)butyl]phenyl beta-D-glucopyranosiduronic acid; 2-{6-[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl]-2-methyl-3,3-dioxido-7-oxo-8-oxa-3lambda6-thia-2,6- diazanonan-9-yl}-5-(4-{[(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetyl]amino}butyl)phenyl beta-D- glucopyranosiduronic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-(1-{[3-(2- {({[2-{[(2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2-yl]oxy}-4-(4-{[(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetyl]amino}butyl)benzyl]oxy}carbonyl)[3-(dimethylamino)-3- oxopropyl]amino}ethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H-pyrazol-4- yl)pyridine-2-carboxylic acid;
2-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2-sulfamoylethyl)carbamoyl}oxy)methyl]-5-(4-{[(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetyl]amino}butyl)phenyl beta-D-glucopyranosiduronic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-(1-{[3-(2- {({[2-{[(2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2-yl]oxy}-4-(4-{[(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetyl]amino}butyl)benzyl]oxy}carbonyl)[3-(methylamino)-3- oxopropyl]amino}ethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H-pyrazol-4- yl)pyridine-2-carboxylic acid;
3-{1-[(3-{2-[(3-amino-3-oxopropyl)({[2-{[(2S,3R,4S,5S,6S)-6-carboxy-3,4,5- trihydroxytetrahydro-2H-pyran-2-yl]oxy}-4-(4-{[(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)acetyl]amino}butyl)benzyl]oxy}carbonyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl)methyl]-5-methyl-1H-pyrazol-4-yl}-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]pyridine-2-carboxylic acid;
2-[({[2-({3-[(4-{6-[3-(1,3-benzothiazol-2-ylcarbamoyl)-1H-indol-5-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-5-(4-{[(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)acetyl]amino}butyl)phenyl beta-D-glucopyranosiduronic acid;
2-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-5,6-dihydroimidazo[1,5- a]pyrazin-7(8H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl]carbamoyl}oxy)methyl]-5-(4-{[(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)acetyl]amino}butyl)phenyl beta-D-glucopyranosiduronic acid;
(6S)-2,6-anhydro-6-(2-{2-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-5,6- dihydroimidazo[1,5-a]pyrazin-7(8H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]- 5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl]carbamoyl}oxy)methyl]-5-({N-[(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)acetyl]-L-valyl-L-alanyl}amino)phenyl}ethyl)-L-gulonic acid; (6S)-2,6-anhydro-6-[2-(2-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5- methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1- yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2- methoxyethyl)carbamoyl}oxy)methyl]-5-{[N-({(3S,5S)-3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)-2- oxo-5-[(2-sulfoethoxy)methyl]pyrrolidin-1-yl}acetyl)-L-valyl-L-alanyl]amino}phenyl)ethyl]-L- gulonic acid;
8-[2-({[(3-amino-3-oxopropyl){2-[(3-{[4-(6-{8-[(1,3-benzothiazol-2-yl)carbamoyl]- 3,4-dihydroisoquinolin-2(1H)-yl}-2-carboxypyridin-3-yl)-5-methyl-1H-pyrazol-1-yl]methyl}-5,7- dimethyltricyclo[3.3.1.13,7]decan-1-yl)oxy]ethyl}carbamoyl]oxy}methyl)-5-{[(2S)-2-({(2S)-2-[2-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetamido]-3-methylbutanoyl}amino)propanoyl]amino}phenyl]- 2,6-anhydro-7,8-dideoxy-L-glycero-L-gulo-octonic acid;
4-{[({2-[(3-{[4-(6-{8-[(1,3-benzothiazol-2-yl)carbamoyl]-3,4-dihydroisoquinolin- 2(1H)-yl}-2-carboxypyridin-3-yl)-5-methyl-1H-pyrazol-1-yl]methyl}-5,7- dimethyltricyclo[3.3.1.13,7]decan-1-yl)oxy]ethyl}[3-(methylamino)-3- oxopropyl]carbamoyl)oxy]methyl}-3-{3-[2-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)acetamido]propoxy}phenyl beta-D-glucopyranosiduronic acid;
2,6-anhydro-8-(2-{[({2-[(3-{[4-(6-{8-[(1,3-benzothiazol-2-yl)carbamoyl]-3,4- dihydroisoquinolin-2(1H)-yl}-2-carboxypyridin-3-yl)-5-methyl-1H-pyrazol-1-yl]methyl}-5,7- dimethyltricyclo[3.3.1.13,7]decan-1-yl)oxy]ethyl}[3-(methylamino)-3- oxopropyl]carbamoyl)oxy]methyl}-5-{[(2S)-2-({(2S)-2-[2-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)acetamido]-3-methylbutanoyl}amino)propanoyl]amino}phenyl)-7,8-dideoxy-L-glycero-L-gulo- octonic acid;
2,6-anhydro-8-(2-{[({2-[(3-{[4-(6-{8-[(1,3-benzothiazol-2-yl)carbamoyl]-3,4- dihydroisoquinolin-2(1H)-yl}-2-carboxypyridin-3-yl)-5-methyl-1H-pyrazol-1-yl]methyl}-5,7- dimethyltricyclo[3.3.1.13,7]decan-1-yl)oxy]ethyl}[3-(methylamino)-3- oxopropyl]carbamoyl)oxy]methyl}-5-{[(2S)-2-{[(2S)-2-(2-{(3S,5S)-3-(2,5-dioxo-2,5-dihydro-1H- pyrrol-1-yl)-2-oxo-5-[(2-sulfoethoxy)methyl]pyrrolidin-1-yl}acetamido)-3- methylbutanoyl]amino}propanoyl]amino}phenyl)-7,8-dideoxy-L-glycero-L-gulo-octonic acid;
6-{8-[(1,3-benzothiazol-2-yl)carbamoyl]-3,4-dihydroisoquinolin-2(1H)-yl}-3-[1-({3- [2-({[(4-{[(2S)-5-(carbamoylamino)-2-{[(2S)-2-{[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl]amino}-3- methylbutanoyl]amino}pentanoyl]amino}phenyl)methoxy]carbonyl}amino)acetamido]-5,7- dimethyltricyclo[3.3.1.13,7]decan-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid; and
8-[2-({[(3-amino-3-oxopropyl){2-[(3-{[4-(6-{8-[(1,3-benzothiazol-2-yl)carbamoyl]- 3,4-dihydroisoquinolin-2(1H)-yl}-2-carboxypyridin-3-yl)-5-methyl-1H-pyrazol-1-yl]methyl}-5,7- dimethyltricyclo[33113,7]decan-1-yl)oxy]ethyl}carbamoyl]oxy}methyl)-5-{[(2S)-2-{[(2S)-2-(2- {(3S,5S)-3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)-2-oxo-5-[(2-sulfoethoxy)methyl]pyrrolidin-1- yl}acetamido)-3-methylbutanoyl]amino}propanoyl]amino}phenyl]-2,6-anhydro-7,8-dideoxy-L- glycero-L-gulo-octonic acid.
In certain embodiments, the ADC, or a pharmaceutically acceptable salt thereof, comprises D is the Bcl-xL inhibitor selected from the group consisting of the following compounds modified in that the hydrogen corresponding to the # position is not present, forming a monoradical:
W3.01, W3.02, W3.03, W3.04, W3.05, W3.06, W3.07, W3.08, W3.09, W3.10, W3.11, W3.12, W3.13, W3.14, W3.15, W3.16, W3.17, W3.18, W3.19, W3.20, W3.21, W3.22, W3.23, W3.24, W3.25, W3.26, W3.27, W3.28, W3.29, W3.30, W3.31, W3.32, W3.33, W3.34, W3.35, W3.36, W3.37, W3.38, W3.39, W3.40, W3.41, W3.42, and W3.43 and pharmaceutically acceptable salts thereof;
L is selected from the group consisting of linkers IVa.1-IVa.4, IVa.8, IVb.1-IVb.13, IVb.15- IVb.19, IVc.1-IVc.7, IVd.1-IVd.4, Va.1-Va.7, Va.10-Va.12, Vb.1-Vb.10, Vc.1-Vc.11, Vd.1-Vd.3, Vd.5-Vd.6, Ve.1-Ve.2, VIa.1, VId.1-VId.2, VId.4, VIIa.1-VIIa.4, VIIb.1-VIIb.8, VIIc.1-VIIc.6 wherein the maleimide of each linker has reacted with the antibody, Ab, forming a covalent attachment as either a succinimide (closed form) or succinamide (open form);
LK is selected from the group consisting of amide, thiourea and thioether; and
m is an integer ranging from 1 to 8.
In certain embodiments, the ADC, or a pharmaceutically acceptable salt thereof,
D is the Bcl-xL inhibitor selected from the group consisting of the following compounds modified in that the hydrogen corresponding to the # position is not present, forming a monoradical:
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-5,6-dihydroimidazo[1,5-a]pyrazin-7(8H)- yl]pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-3-{1- [(3-{2-[(2-methoxyethyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl- 1H-pyrazol-4-yl}pyridine-2-carboxylic acid;
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-cyano-3,4-dihydroisoquinolin-2(1H)- yl]pyridine-2-carboxylic acid;
6-[4-(1,3-benzothiazol-2-ylcarbamoyl)isoquinolin-6-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid; 3-{1-[(3-{2-[(3-amino-3-oxopropyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl)methyl]-5-methyl-1H-pyrazol-4-yl}-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]pyridine-2-carboxylic acid;
and pharmaceutically acceptable salts thereof;
L is selected from the group consisting of linkers IVb.2, IVc.5, IVc.6, IVc.7, Vc.11, IVd.4, Vb.9, VIIa.1, VIIa.3, VIIc.1, VIIc.4, and VIIc.5 in either closed or open forms, and pharmaceutically acceptable salts thereof;
LK is thioether; and
m is an integer ranging from 2 to 4.
To form an ADC, the maleimide ring of a synthon (for example, the synthons listed in Table B) may react with an antibody Ab, forming a covalent attachment as either a succinimide (closed form) or succinamide (open form). Similarly, other functional groups, e.g. acetyl halide or vinyl sulfone may react with an antibody, Ab, forming a covalent attachment.
In certain embodiments, the ADC, or a pharmaceutically acceptable salt thereof, is selected from the group consisting of huAb13v1-ZT, huAb13v1-ZZ, huAb13v1-XW, huAb13v1-SE, huAb13v1-SR, huAb13v1-YG, huAb13v1-KZ, huAb3v2.5-ZT, huAb3v2.5-ZZ, huAb3v2.5-XW, huAb3v2.5-SE, huAb3v2.5-SR, huAb3v2.5-YG, huAb3v2.5-KZ, huAb3v2.6-ZT, huAb3v2.6-ZZ, huAb3v2.6-XW, huAb3v2.6-SE, huAb3v2.6-SR, huAb3v2.6-YG, and huAb3v2.6-KZ, wherein KZ, SR, SE, XW, YG, ZT and ZZ are synthons disclosed in Table B, and wherein the conjugated synthons are either in open or closed form. In a specific embodiment, the ADC is huAb13v1-ZT, huAb13v1- ZZ, huAb13v1-XW, huAb13v1-SE, huAb13v1-SR, huAb13v1-YG, huAb13v1-KZ, huAb3v2.5-ZT, huAb3v2.5-ZZ, huAb3v2.5-XW, huAb3v2.5-SE, huAb3v2.5-SR, huAb3v2.5-YG, huAb3v2.5-KZ, huAb3v2.6-ZT, huAb3v2.6-ZZ, huAb3v2.6-XW, huAb3v2.6-SE, huAb3v2.6-SR, huAb3v2.6-YG, and huAb3v2.6-KZ, wherein huAb13v1, huAb3v2.5, and huAb3v2.6 are the anti-hB7-H3 antibodies and KZ, SR, SE, XW, YG, ZT and ZZ are synthons disclosed in Table B, and wherein the conjugated synthons are either in open or closed form.
In certain embodiments, the ADC, or a pharmaceutically acceptable salt thereof, is
Figure imgf000218_0001
Figure imgf000219_0001
Figure imgf000220_0001
Figure imgf000221_0001
Figure imgf000222_0001
wherein m is 2, Ab is either an anti-hB7-H3 antibody, wherein the anti-hB7-H3 antibody comprises a heavy chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 12, a heavy chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 140, and a heavy chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 10; and a light chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 15, a light chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 7, and a light chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 136; or an anti-hB7-H3 antibody, wherein the anti-hB7H3 antibody comprises a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 139, and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 135; or an anti-hB7-H3 antibody, wherein the anti-hB7-H3 antibody comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 170, and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 171.
In one embodiment, the ADC, or a pharmaceutically acceptable salt thereof, is
Figure imgf000222_0002
wherein m is 2, Ab is either an anti-hB7-H3 antibody, wherein the anti-hB7-H3 antibody comprises a heavy chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 35, a heavy chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 34, and a heavy chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 33; and a light chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 39, a light chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 38, and a light chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 37; or an anti-hB7-H3 antibody, wherein the anti-hB7H3 antibody comprises a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 147, and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 144; or an anti-hB7-H3 antibody, wherein the anti-hB7-H3 antibody comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 168, and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 169.
Figure imgf000223_0001
wherein m is 2, Ab is either an anti-hB7-H3 antibody, wherein the anti-hB7-H3 antibody comprises a heavy chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 12, a heavy chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 140, and a heavy chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 10; and a light chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 15, a light chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 7, and a light chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 136; or an anti-hB7-H3 antibody, wherein the anti-hB7H3 antibody comprises a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 139, and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 135; or an anti-hB7-H3 antibody, wherein the anti-hB7-H3 antibody comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 170, and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 171.
Figure imgf000224_0001
wherein m is 2, Ab is either an anti-hB7-H3 antibody, wherein the anti-hB7-H3 antibody comprises a heavy chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 35, a heavy chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 34, and a heavy chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 33; and a light chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 39, a light chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 38, and a light chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 37; or an anti-hB7-H3 antibody, wherein the anti-hB7H3 antibody comprises a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 147, and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 144; or an anti-hB7-H3 antibody, wherein the anti-hB7- H3 antibody comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 168, and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 169.
Bcl-xL inhibitors, including warheads and synthons, and methods of making the same are described in US 2016-0158377 (AbbVie Inc.), which is incorporated by reference herein. III.A.4. Methods of Synthesis of Bcl-xL ADCs
The Bcl-xL inhibitors and synthons described herein may be synthesized using standard, known techniques of organic chemistry. General schemes for synthesizing Bcl-xL inhibitors and synthons that may be used as-is or modified to synthesize the full scope of Bcl-xL inhibitors and synthons described herein are provided below. Specific methods for synthesizing exemplary Bcl-xL inhibitors and synthons that may be useful for guidance are provided in the Examples section.
ADCs may likewise be prepared by standard methods, such as methods analogous to those described in Hamblett et al., 2004,“Effects of Drug Loading on the Antitumor Activity of a
Monoclonal Antibody Drug Conjugate”, Clin. Cancer Res.10:7063-7070; Doronina et al., 2003, “Development of potent and highly efficacious monoclonal antibody auristatin conjugates for cancer therapy,” Nat. Biotechnol.21(7):778-784; and Francisco et al., 2003,“cAClO-vcMMAE, an anti- CD30-monomethylauristatin E conjugate with potent and selective antitumor activity,” Blood 102:1458-1465. For example, ADCs with four drugs per antibody may be prepared by partial reduction of the antibody with an excess of a reducing reagent such as DTT or TCEP at 37 °C for 30 min, then the buffer exchanged by elution through SEPHADEX® G-25 resin with 1 mM DTPA in DPBS. The eluent is diluted with further DPBS, and the thiol concentration of the antibody may be measured using 5,5’-dithiobis(2-nitrobenzoic acid) [Ellman’s reagent]. An excess, for example 5- fold, of a linker-drug synthon is added at 4 °C for 1 hour, and the conjugation reaction may be quenched by addition of a substantial excess, for example 20-fold, of cysteine. The resulting ADC mixture may be purified on SEPHADEX G-25 equilibrated in PBS to remove unreacted synthons, desalted if desired, and purified by size-exclusion chromatography. The resulting ADC may then be then sterile-filtered, for example, through a 0.2 µm filter, and lyophilized if desired for storage. In certain embodiments, all of the interchain cysteine disulfide bonds are replaced by linker-drug conjugates. One embodiment pertains to a method of making an ADC, comprising contacting a synthon described herein with an antibody under conditions in which the synthon covalently links to the antibody.
Specific methods for synthesizing exemplary ADCs that may be used to synthesize the full range of ADCs described herein are provided in the Examples section.
III.A.5. General Methods for Synthesizing Bcl-xL Inhibitors
In the schemes below, the various substituents Ar1, Ar2, Z1, R4, R10, R11a and R11b are as defined in the Detailed Description section.
5.1.1 Synthesis of Compound (9)
Figure imgf000226_0001
T e synt es s o compoun 9 s escr e n Sc eme 1. Compoun 1 can e treate with BH3·THF to afford compound (2). The reaction is typically performed at ambient temperature in a solvent, such as, but not limited to, tetrahydrofuran. Compound (3) can be prepared by treating compound (2) with
Figure imgf000226_0002
in the presence of cyanomethylenetributylphosphorane. The reaction is typically performed at an elevated temperature in a solvent such as, but not limited to, toluene.
Compound (3) can be treated with ethane-1,2-diol in the presence of a base such as, but not limited to, triethylamine, to provide compound (4). The reaction is typically performed at an elevated temperature, and the reaction may be performed under microwave conditions. Compound (4) can be treated with a strong base, such as, but not limited to, n-butyllithium, followed by the addition of iodomethane, to provide compound (5). The addition and reaction is typically performed in a solvent such as, but not limited to, tetrahydrofuran, at a reduced temperature before warming up to ambient temperature for work up. Compound (5) can be treated with N-iodosuccinimide to provide compound (6). The reaction is typically performed at ambient temperature is a solvent such as, but not limited to, N,N-dimethylformamide. Compound (7) can be prepared by reacting compound (6) with methanesulfonyl chloride, in the presence of a base such as, but not limited to, triethylamine, followed by the addition of NHR4. The reaction with methanesulfonyl chloride is typically performed at low temperature, before increasing the temperature for the reaction with NHR4, and the reaction is typically performed in a solvent such as, but not limited to tetrahydrofuran. Compound (7) can be reacted with di-tert-butyl dicarbonate in the presence of 4-dimethylaminopyridine to provide compound (8). The reaction is typically performed at ambient temperature in a solvent such as, but not limited to tetrahydrofuran. The borylation of compound (8) to provide compound (9) can be performed under conditions described herein and readily available in the literature.
5.1.2. Synthesis of Compound (12)
Figure imgf000227_0001
The synthesis of intermediate (12) is described in Scheme 2. Compound (3) can be treated with tri-n-butyl-allylstannane in the presence of ZnCl2·Et2O or N, N'-azoisobutyronitrile (AIBN) to provide compound (10) (Yamamoto et al., 1998, Heterocycles 47:765-780). The reaction is typically performed at -78 °C in a solvent, such as, but not limited to dichloromethane. Compound (10) can be treated under standard conditions known in the art for hydroboration/oxidation to provide compound (11). For example, treatment of compound (10) with a reagent such as BH3·THF in a solvent such as, but not limited to, tetrahydrofuran followed by treatment of the intermediate alkylborane adduct with an oxidant such as, but not limited to, hydrogen peroxide in the presence of a base such as, but not limited to, sodium hydroxide would provide compound (11) (Brown et al., 1968, J. Am. Chem. Soc., 86:397). Typically the addition of BH3·THF is performed at low temperature before warming to ambient temperature, which is followed by the addition of hydrogen peroxide and sodium hydroxide to generate the alcohol product. Compound (12) can be generated according to Scheme 1, as previously described for compound (9). 5.1.3. Synthesis of Compound (15)
Scheme 3
The synthesis of intermediate (15), is described in Scheme 3. Compound (3) can be reacted with thiourea in a solvent mixture of acetic acid and 48% aqueous HBr solution at 100 °C to yield an intermediate that can be subsequently treated with sodium hydroxide in a solvent mixture such as, but not limited to, 20% v/v ethanol in water to provide compound (13). Compound (13) can be reacted with 2-chloroethanol in the presence of a base such as, but not limited to, sodium ethoxide to provide compound (14). The reaction is typically performed at ambient or elevated temperatures in a solvent such as, but not limited to, ethanol. Compound (15) can be generated according to Scheme 1, as previously described for compound (9).
.1
Figure imgf000229_0001
v
44 92 99 24 E1 The synthesis of compound (22) is described in Scheme 4. Compound (16) can be reacted with iodomethane in the presence of a base such as, but not limited to, potassium carbonate to provide compound (17). The reaction is typically conducted at ambient or elevated temperature in a solvent such as, but not limited to, acetone or N,N-dimethylformamide. Compound (17) can be reacted under photochemical conditions with tosyl cyanide in the presence of benzophenone to provide compound (18) (see Kamijo et al., Org. Lett., 2011, 13:5928-5931). The reaction is typically run at ambient temperature in a solvent such as, but not limited to, acetonitrile or benzene using a Riko 100W medium pressure mercury lamp as the light source. Compound (18) can be reacted with lithium hydroxide in a solvent system such as, but not limited to, mixtures of water and tetrahydrofuran or water and methanol to provide compound (19). Compound (19) can be treated with BH3·THF to provide compound (20). The reaction is typically performed at ambient temperature in a solvent, such as, but not limited to, tetrahydrofuran. Compound (21) can be prepared by treating compound (20)
with
Figure imgf000230_0001
in the presence of cyanomethylenetributylphosphorane. The reaction is typically performed at an elevated temperature in a solvent such as, but not limited to, toluene. Compound (21) can be treated with N-iodosuccinimide to provide compound (22). The reaction is typically performed at ambient temperature is a solvent such as, but not limited to, N,N-dimethylformamide.
5.1.5. Synthesis of Compound (24)
Figure imgf000230_0002
The synthesis of compound (24) is described in Scheme 5. Compound (22) can be treated with a reducing agent such as, but not limited to, lithium aluminum hydride in a solvent such as, but not limited to, diethyl ether or tetrahydrofuran to provide compound (23). Typically the reaction is performed at 0 °C before warming to ambient or elevated temperature. Compound (23) can be reacted with di-tert-butyl dicarbonate under standard conditions described herein or in the literature to provide compound (24).
5.1.6. Synthesis of Compound (24a)
Figure imgf000230_0003
The synthesis of intermediate (24a) is described in Scheme 6. Compound (22a) can be hydrolyzed using conditions described in the literature to provide compound (23a). Typically the reaction is run in the presence of potassium hydroxide in a solvent such as, but not limited to, ethylene glycol at elevated temperatures (see Roberts et al., 1994, J. Org. Chem., 1994, 59:6464-6469; Yang et al, 2013, Org. Lett., 15:690-693). Compound (24a) can be made from compound (23a) by Curtius rearrangement using conditions described in the literature. For example, compound (23a) can be reacted with sodium azide in the presence of tetrabutylammonium bromide, zinc(II) triflate and di- tert-butyl dicarbonate to provide compound (24a) (see Lebel et al., Org. Lett., 2005, 7:4107-4110). Typically the reaction is run at elevated temperatures, preferably from 40-50 °C, in a solvent such as, but not limited to, tetrahydrofuran.
5.1.7. Synthesis of Compound (29)
Figure imgf000231_0001
Scheme 7 describes a functionalization of the adamantane ring substituent. Dimethyl sulfoxide can be reacted with oxalyl chloride, followed by the addition of compound (25), in the presence of a base such as, but not limited to triethylamine, to provide compound (26). The reaction is typically performed at low temperature in a solvent such as, but not limited to, dichloromethane. Compound (27) can be reacted with compound (26), followed by treatment with sodium borohydride, to provide compound (28). The reaction is typically performed at ambient temperature in a solvent such as, but not limited to, dichloromethane, methanol, or mixtures thereof. Compound (29) can be prepared by reacting compound (28) with di-tert-butyl dicarbonate, in the presence of N,N- dimethylpyridin-4-amine. The reaction is typically performed at ambient temperature in a solvent such as, but not limited to, tetrahydrofuran. 5.1.8. Synthesis of Compound (35)
Scheme 8
Figure imgf000232_0001
As shown in Scheme 8, compound (30), can be reacted with compound (31) under Suzuki coupling conditions described herein and readily available in the literature, to provide compound (32). Compound (34) can be prepared by reacting compound (32) with compound (33) under conditions described herein, and readily available in the literature. Compound (35) can be prepared by treating compound (34) with an acid such as, but not limited to, trifluoroacetic acid. The reaction is typically performed at ambient temperature in a solvent such as, but not limited to, dichloromethane.
5.1.9. Synthesis of Compound (43)
Scheme 9
Figure imgf000232_0002
Scheme 9 describes the synthesis of substituted 1,2,3,4-tetrahydroisoquinoline intermediates. Trimethylsilanecarbonitrile can be treated with tetrabutylammonium fluoride and then reacted with compound (36), wherein X is Br or I, to provide compound (37). The additions are typically performed at ambient temperature before heating to an elevated temperature, in a solvent such as, but not limited to, tetrahydrofuran, acetonitrile, or mixtures thereof. Compound (37) can be treated with borane to provide compound (38). The reaction is typically performed at ambient temperature in a solvent such as, but not limited to, tetrahydrofuran. Compound (39) can be prepared by treating compound (38) with trifluoroacetic anhydride, in the presence of a base such as, but no limited to, triethylamine. The reaction is initially performed at low temperature before warming to ambient temperature in a solvent such as, but not limited to, dichloromethane. Compound (39) can be treated with paraformaldehyde in the presence of sulfuric acid to provide compound (40). The reaction is typically performed at ambient temperature. Compound (41) can be prepared by reacting compound (40) with dicyanozinc in the presence of a catalyst such as, but not limited to,
tetrakis(triphenylphosphine)palladium(0). The reaction is typically performed at an elevated temperature under a nitrogen atmosphere in a solvent such as, but not limited to,
N,N-dimethylformamide. Compound (41) can be treated with potassium carbonate to provide compound (42). The reaction is typically performed at ambient temperature in a solvent such as, but not limited to, methanol, tetrahydrofuran, water, or mixtures thereof.
5.1.10 Synthesis of Compound (47)
Figure imgf000233_0001
As shown in Scheme 10, compound (45) can be prepared by reacting compound (43), with tert-butyl 3-bromo-6-fluoropicolinate (44) in the presence of a base, such as, but not limited to, N,N-diisopropylethylamine or triethylamine. The reaction is typically performed under an inert atmosphere at an elevated temperature, in a solvent, such as, but not limited to, dimethyl sulfoxide. Compound (45) can be reacted with 4,4,5,5-tetramethyl-1,3,2-dioxaborolane (46), under borylation conditions described herein or in the literature to provide compound (47). 5.1.11. Synthesis of Compound (53)
Figure imgf000234_0001
Scheme 11 describes the synthesis of optionally substituted 1,2,3,4-tetrahydroisoquinoline Bcl-xL inhibitors. Compound (47) can be prepared by reacting compound (45) with pinacolborane, in the presence of a base such as but not limited to triethylamine, and a catalyst such as but not limited to [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II). The reaction is typically performed at an elevated temperature in a solvent such as, but not limited to acetonitrile. Compound (50) can be prepared by reacting compound (47) with compound (8) under Suzuki coupling conditions described herein and readily available in the literature. Compound (50) can be treated with lithium hydroxide to provide compound (51). The reaction is typically performed at ambient temperature in a solvent such as, but not limited to, tetrahydrofuran, methanol, water, or mixtures thereof. Compound (51) can be reacted with compound (33) under amidation conditions described herein and readily available in the literature to provide compound (52). Compound (53) can be prepared by treating compound (52) with an acid such as, but not limited to, trifluoroacetic acid. The reaction is typically performed at ambient temperature in a solvent such as, but not limited to, dichloromethane. 5.1.12. Synthesis of Compound (66)
Scheme 12
Figure imgf000235_0001
Scheme 12 describes the synthesis of 5-methoxy 1,2,3,4-tetrahydroisoquinoline Bcl-xL inhibitors. tert-Butyl 8-bromo-5-hydroxy-3,4-dihydroisoquinoline-2(1H)-carboxylate (54) can be prepared by treating tert-butyl 5-hydroxy-3,4-dihydroisoquinoline-2(1H)-carboxylate with N- bromosuccinimide. The reaction is typically performed at ambient temperature in a solvent such as, but not limited to N,N-dimethylformamide. Butyl 8-bromo-5-hydroxy-3,4-dihydroisoquinoline- 2(1H)-carboxylate (54) can be reacted with benzyl bromide (55) in the presence of a base such as, but not limited to, potassium carbonate to provide tert-butyl 5-(benzyloxy)-8-bromo-3,4- dihydroisoquinoline-2(1H)-carboxylate (56). The reaction is typically performed at an elevated temperature in a solvent such as, but not limited to, acetone. tert-Butyl 5-(benzyloxy)-8-bromo-3,4- dihydroisoquinoline-2(1H)-carboxylate (56) can be treated with carbon monoxide in the presence of methanol and a base such as, but not limited to, triethylamine, and a catalyst such as but not limited to[1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II), to provide 2-tert-butyl 8-methyl 5- (benzyloxy)-3,4-dihydroisoquinoline-2,8(1H)-dicarboxylate (57). The reaction is typically performed at an elevated temperature. Methyl 5-(benzyloxy)-1,2,3,4-tetrahydroisoquinoline-8-carboxylate (58) can be prepared by treating 2-tert-butyl 8-methyl 5-(benzyloxy)-3,4-dihydroisoquinoline-2,8(1H)- dicarboxylate (57) with hydrochloric acid. The reaction is typically performed at ambient temperature, in a solvent such as, but not limited to, tetrahydrofuran, dioxane, or mixtures thereof. Methyl 5-(benzyloxy)-1,2,3,4-tetrahydroisoquinoline-8-carboxylate (58) can be reacted with tert-butyl 3-bromo-6-fluoropicolinate (44) in the presence of a base such as, but not limited to, triethylamine, to provide methyl 5-(benzyloxy)-2-(5-bromo-6-(tert-butoxycarbonyl)pyridin-2-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxylate (59). The reaction is typically performed at elevated temperature in a solvent such as, but not limited to, dimethyl sulfoxide. Methyl 5-(benzyloxy)-2-(5- bromo-6-(tert-butoxycarbonyl)pyridin-2-yl)-1,2,3,4-tetrahydroisoquinoline-8-carboxylate (59) can be reacted with compound (60), wherein Ad is a methyladamantane moiety of the compounds of the disclosure (e.g., the compounds of formula (IIa) and (IIb)) under Suzuki coupling conditions described herein and readily available in the literature, to provide compound (61). Compound (61) can be treated with hydrogen gas in the presence of palladium hydroxide to provide compound (62). The reaction is typically performed at elevated temperature in a solvent such as, but not limited to, tetrahydrofuran. Compound (63) can be prepared by reacting compound (62) with
(trimethylsilyl)diazomethane. The reaction is typically performed at ambient temperature, in a solvent such as, but not limited to, dichloromethane, methanol, diethyl ether, or mixtures thereof. Compound (63) can be treated with lithium hydroxide to provide compound (64). The reaction is typically performed at ambient temperature in a solvent such as, but not limited to, tetrahydrofuran, methanol, water, or mixtures thereof. Compound (64) can be reacted with compound (33) under amidation conditions described herein and readily available in the literature to provide compound (65). Compound (66) can be prepared by treating compound (65) with hydrochloric acid. The reaction is typically performed at ambient temperature in a solvent such as, but not limited to, dioxane.
III.A.6. General Methods for Synthesizing Synthons
In the schemes below, the various substituents Ar1, Ar2, Z1, R4, R11a and R11b are as defined in the Detailed Description section.
1 v. 44 92
Figure imgf000237_0001
9 9
24 E1 As shown in scheme 13, compounds of formula (77), wherein PG is an appropriate base labile protecting group and AA(2) is Cit, Ala, or Lys, can be reacted with 4-(aminophenyl)methanol (78), under amidation conditions described herein or readily available in the literature to provide compound (79). Compound (80) can be prepared by reacting compound (79) with a base such as, but not limited to, diethylamine. The reaction is typically performed at ambient temperature in a solvent such as but not limited to N,N-dimethylformamide. Compound (81), wherein PG is an appropriate base or acid labile protecting group and AA(1) is Val or Phe, can be reacted with compound (80), under amidation conditions described herein or readily available in the literature to provide compound (82).
Compound (83) can be prepared by treating compound (82) with diethylamine or trifluoroacetic acid, as appropriate. The reaction is typically performed at ambient temperature in a solvent such as but not limited to dichloromethane. Compound (84), wherein Sp is a spacer, can be reacted with compound (83) to provide compound (85). The reaction is typically performed at ambient temperature in a solvent such as but not limited to N,N-dimethylformamide. Compound (85) can be reacted with bis(4-nitrophenyl) carbonate (86) in the presence of a base such as, but not limited to N,N-diisopropylethylamine, to provide compounds (87). The reaction is typically performed at ambient temperature in a solvent such as but not limited to N,N-dimethylformamide. Compounds (87) can be reacted with compounds of formula (88) in the presence of a base such as, but not limited to, N,N-diisopropylethylamine, to provide compound (89). The reaction is typically performed at ambient temperature in a solvent such as, but not limited to, N,N-dimethylformamide.
.1 v 44 92 99 24
Figure imgf000239_0001
E 1 Scheme 14 describes the installment of alternative mAb-linker attachments to dipeptide synthons. Compound (88), wherein can be reacted with compound (90) in the presence of a base such as, but not limited to, N-ethyl-N-isopropylpropan-2-amine, to provide compound (91). The reaction is typically performed at ambient temperature in a solvent such as but not limited to
N,N-dimethylformamide. Compound (92) can be prepared by reacting compound (91) with diethylamine. The reaction is typically performed at ambient temperature in a solvent such as but not limited to N,N-dimethylformamide. Compound (93), wherein X1 is Cl, Br, or I, can be reacted with compound (92), under amidation conditions described herein or readily available in the literature to provide compound (94). Compound (92) can be reacted with compounds of formula (95) under amidation conditions described herein or readily available in the literature to provide compound (96).
.1 v 44 92
Figure imgf000241_0001
9 9
24 E1 Scheme 15 describes the synthesis of vinyl glucuronide linker intermediates and synthons. (2R,3R,4S,5S,6S)-2-Bromo-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (97) can be treated with silver oxide, followed by 4-bromo-2-nitrophenol (98) to provide (2S,3R,4S,5S,6S)-2- (4-bromo-2-nitrophenoxy)-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (99). The reaction is typically performed at ambient temperature in a solvent, such as, but not limited to, acetonitrile. (2S,3R,4S,5S,6S)-2-(4-Bromo-2-nitrophenoxy)-6-(methoxycarbonyl)tetrahydro-2H- pyran-3,4,5-triyl triacetate (99) can be reacted with (E)-tert-butyldimethyl((3-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)allyl)oxy)silane (100) in the presence of a base such as, but not limited to, sodium carbonate, and a catalyst such as but not limited to tris(dibenzylideneacetone)dipalladium (Pd2(dba)3), to provide (2S,3R,4S,5S,6S)-2-(4-((E)-3-((tert-butyldimethylsilyl)oxy)prop-1-en-1-yl)- 2-nitrophenoxy)-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (101). The reaction is typically performed at an elevated temperature in a solvent, such as, but not limited to,
tetrahydrofuran. (2S,3R,4S,5S,6S)-2-(2-amino-4-((E)-3-hydroxyprop-1-en-1-yl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (102) can be prepared by reacting (2S,3R,4S,5S,6S)-2-(4-((E)-3-((tert-butyldimethylsilyl)oxy)prop-1-en-1-yl)-2-nitrophenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (101) with zinc in the presence of an acid such as, but not limited to, hydrochloric acid. The addition is typically performed at low temperature before warming to ambient temperature in a solvent such as, but not limited to, tetrahydrofuran, water, or mixtures thereof. (2S,3R,4S,5S,6S)-2-(2-amino-4-((E)-3-hydroxyprop-1-en-1-yl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (102) can be reacted with (9H-fluoren-9- yl)methyl (3-chloro-3-oxopropyl)carbamate (103), in the presence of a base such as, but not limited to, N,N-diisopropylethylamine, to provide (2S,3R,4S,5S,6S)-2-(2-(3-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)propanamido)-4-((E)-3-hydroxyprop-1-en-1-yl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (104). The addition is typically performed at low temperature before warming to ambient temperature in a solvent such as, but not limited to, dichloromethane. Compound (88) can be reacted with (2S,3R,4S,5S,6S)-2-(2-(3-((((9H- fluoren-9-yl)methoxy)carbonyl)amino)propanamido)-4-((E)-3-hydroxyprop-1-en-1-yl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (104) in the presence of a base such as, but not limited to, N-ethyl-N-isopropylpropan-2-amine, followed by work up and reaction with compound (105) in the presence of a base such as, but not limited to, N,N-diisopropylethylamine to provide compound (106). The reactions are typically performed at ambient temperature in a solvent such as, but not limited to N,N- dimethylformamide.
2
16
e
em
h
Sc
5)
(11
nd
u
po
om
C
of
sis
he
nt
Sy 4 .
2.
5.
Figure imgf000243_0001
.1 v 44 92 99 24 E1 Scheme 16 describes the synthesis of a representative 2-ether glucuronide linker intermediate and synthon. (2S,3R,4S,5S,6S)-2-Bromo-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (97) can be reacted with 2,4-dihydroxybenzaldehyde (107) in the presence of silver carbonate to provide (2S,3R,4S,5S,6S)-2-(4-formyl-3-hydroxyphenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (108). The reaction is typically performed at an elevated temperature in a solvent, such as, but not limited to, acetonitrile.
(2S,3R,4S,5S,6S)-2-(4-Formyl-3-hydroxyphenoxy)-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5- triyl triacetate (108) can be treated with sodium borohydride to provide (2S,3R,4S,5S,6S)-2-(3- hydroxy-4-(hydroxymethyl)phenoxy)-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (109). The addition is typically performed at low temperature before warming to ambient temperature in a solvent such as but not limited to tetrahydrofuran, methanol, or mixtures thereof.
(2S,3R,4S,5S,6S)-2-(4-(((tert-butyldimethylsilyl)oxy)methyl)-3-hydroxyphenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (110) can be prepared by reacting (2S,3R,4S,5S,6S)-2-(3-hydroxy-4-(hydroxymethyl)phenoxy)-6-(methoxycarbonyl)tetrahydro-2H- pyran-3,4,5-triyl triacetate (109) with tert-butyldimethylsilyl chloride in the presence of imidazole. The reaction is typically performed at low temperature in a solvent, such as, but not limited to, dichloromethane. (2S,3R,4S,5S,6S)-2-(3-(2-(2-((((9H-Fluoren-9- yl)methoxy)carbonyl)amino)ethoxy)ethoxy)-4-(((tert-butyldimethylsilyl)oxy)methyl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (111) can be prepared by reacting (2S,3R,4S,5S,6S)-2-(4-(((tert-butyldimethylsilyl)oxy)methyl)-3-hydroxyphenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (110) with (9H-fluoren-9-yl)methyl (2- (2-hydroxyethoxy)ethyl)carbamate in the presence of triphenylphosphine and a azodicarboxylate such as, but not limited to, di-tert-butyl diazene-1,2-dicarboxylate. The reaction is typically performed at ambient temperature in a solvent such as but not limited to toluene. (2S,3R,4S,5S,6S)-2-(3-(2-(2- ((((9H-Fluoren-9-yl)methoxy)carbonyl)amino)ethoxy)ethoxy)-4-(((tert- butyldimethylsilyl)oxy)methyl)phenoxy)-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (111) can be treated with acetic acid to provide (2S,3R,4S,5S,6S)-2-(3-(2-(2-((((9H-fluoren- 9-yl)methoxy)carbonyl)amino)ethoxy)ethoxy)-4-(hydroxymethyl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (112). The reaction is typically performed at ambient temperature in a solvent such as but not limited to water, tetrahydrofuran, or mixtures thereof. (2S,3R,4S,5S,6S)-2-(3-(2-(2-((((9H-Fluoren-9- yl)methoxy)carbonyl)amino)ethoxy)ethoxy)-4-((((4-nitrophenoxy)carbonyl)oxy)methyl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (113) can be prepared by reacting (2S,3R,4S,5S,6S)-2-(3-(2-(2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)ethoxy)ethoxy)-4- (hydroxymethyl)phenoxy)-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (91) with bis(4-nitrophenyl) carbonate in the presence of a base such as but not limited to N-ethyl-N- isopropylpropan-2-amine. The reaction is typically performed at ambient temperature in a solvent such as but not limited to N,N-dimethylformamide. (2S,3R,4S,5S,6S)-2-(3-(2-(2-((((9H-Fluoren-9- yl)methoxy)carbonyl)amino)ethoxy)ethoxy)-4-((((4-nitrophenoxy)carbonyl)oxy)methyl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (113) can be treated with compound (88) in the presence of a base such as but not limited to N-ethyl-N-isopropylpropan-2-amine, followed by treatment with lithium hydroxide to provide a compound (114). The reaction is typically performed at ambient temperature in a solvent such as but not limited to N,N-dimethylformamide, tetrahydrofuran, methanol, or mixtures thereof. Compound (115) can be prepared by reacting compound (114) with compound (84) in the presence of a base such as but not limited to N-ethyl-N-isopropylpropan-2- amine. The reaction is typically performed at ambient temperature in a solvent such as but not limited to N,N-dimethylformamide.
5.2.5. Synthesis of Compound (119)
Scheme 17
Figure imgf000245_0001
Scheme 17 describes the introduction of a second solubilizing group to a sugar linker.
Compound (116) can be reacted with (R)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3- sulfopropanoic acid (117), under amidation conditions described herein or readily available in the literature, followed by treatment with a base such as but not limited to diethylamine, to provide compound (118). Compound (118) can be reacted with compound (84), wherein Sp is a spacer, under amidation conditions described herein or readily available in the literature, to provide compound (119). 5.2.6. Synthesis of Compound (129)
Figure imgf000246_0001
Scheme 18 describes the synthesis of 4-ether glucuronide linker intermediates and synthons. 4-(2-(2-Bromoethoxy)ethoxy)-2-hydroxybenzaldehyde (122) can be prepared by reacting 2,4- dihydroxybenzaldehyde (120) with 1-bromo-2-(2-bromoethoxy)ethane (121) in the presence of a base such as, but not limited to, potassium carbonate. The reaction is typically performed at an elevated temperature in a solvent such as but not limited to acetonitrile. 4-(2-(2-Bromoethoxy)ethoxy)-2- hydroxybenzaldehyde (122) can be treated with sodium azide to provide 4-(2-(2-azidoethoxy)ethoxy)- 2-hydroxybenzaldehyde (123). The reaction is typically performed at ambient temperature in a solvent such as but not limited to N,N-dimethylformamide. (2S,3R,4S,5S,6S)-2-(5-(2-(2- Azidoethoxy)ethoxy)-2-formylphenoxy)-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (125) can be prepared by reacting 4-(2-(2-azidoethoxy)ethoxy)-2-hydroxybenzaldehyde (123) with (3R,4S,5S,6S)-2-bromo-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (124) in the presence of silver oxide. The reaction is typically performed at ambient temperature in a solvent such as, but not limited to, acetonitrile. Hydrogenation of (2S,3R,4S,5S,6S)-2-(5-(2-(2- azidoethoxy)ethoxy)-2-formylphenoxy)-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (125) in the presence of Pd/C will provide (2S,3R,4S,5S,6S)-2-(5-(2-(2- aminoethoxy)ethoxy)-2-(hydroxymethyl)phenoxy)-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5- triyl triacetate (126). The reaction is typically performed at ambient temperature in a solvent such as, but not limited to, tetrahydrofuran. (2S,3R,4S,5S,6S)-2-(5-(2-(2-((((9H-Fluoren-9- yl)methoxy)carbonyl)amino)ethoxy)ethoxy)-2-(hydroxymethyl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (127) can be prepared by treating (2S,3R,4S,5S,6S)-2-(5-(2-(2-aminoethoxy)ethoxy)-2-(hydroxymethyl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (126) with (9H-fluoren-9-yl)methyl carbonochloridate in the presence of a base, such as, but not limited to, N-ethyl-N-isopropylpropan-2- amine. The reaction is typically performed at low temperature in a solvent such as, but not limited to, dichloromethane. Compound (88) can be reacted with (2S,3R,4S,5S,6S)-2-(5-(2-(2-((((9H-Fluoren-9- yl)methoxy)carbonyl)amino)ethoxy)ethoxy)-2-(hydroxymethyl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (127) in the presence of a base, such as, but not limited to, N-ethyl-N-isopropylpropan-2-amine, followed by treatment with lithium hydroxide to provide compound (128). The reaction is typically performed at low temperature in a solvent such as, but not limited to, N,N-dimethylformamide. Compound (129) can be prepared by reacting compound (128) with compound (84) in the presence of a base such as, but not limited to, N-ethyl-N- isopropylpropan-2-amine. The reaction is typically performed at ambient temperature in a solvent such as but not limited to N,N-dimethylformamide.
5.2.7. Synthesis of Compound (139)
Scheme 19
Figure imgf000248_0001
Scheme 19 describes the synthesis of carbamate glucuronide intermediates and synthons. 2- Amino-5-(hydroxymethyl)phenol (130) can be treated with sodium hydride and then reacted with 2- (2-azidoethoxy)ethyl 4-methylbenzenesulfonate (131) to provide (4-amino-3-(2-(2- azidoethoxy)ethoxy)phenyl)methanol (132). The reaction is typically performed at an elevated temperature in a solvent such as, but not limited to N,N-dimethylformamide. 2-(2-(2- Azidoethoxy)ethoxy)-4-(((tert-butyldimethylsilyl)oxy)methyl)aniline (133) can be prepared by reacting (4-amino-3-(2-(2-azidoethoxy)ethoxy)phenyl)methanol (132) with tert- butyldimethylchlorosilane in the presence of imidazole. The reaction is typically performed at ambient temperature in a solvent such as, but not limited to tetrahydrofuran. 2-(2-(2- Azidoethoxy)ethoxy)-4-(((tert-butyldimethylsilyl)oxy)methyl)aniline (133) can be treated with phosgene, in the presence of a base such as but not limited to triethylamine, followed by reaction with (3R,4S,5S,6S)-2-hydroxy-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (134) in the presence of a base such as but not limited to triethylamine, to provide 2S,3R,4S,5S,6S)-2-(((2-(2-(2- azidoethoxy)ethoxy)-4-(((tert-butyldimethylsilyl)oxy)methyl)phenyl)carbamoyl)oxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (135). The reaction is typically performed in a solvent such as, but not limited to, toluene, and the additions are typically performed at low temperature, before warming up to ambient temperature after the phosgene addition and heating at an elevated temperature after the (3R,4S,5S,6S)-2-hydroxy-6-(methoxycarbonyl)tetrahydro-2H- pyran-3,4,5-triyl triacetate (134) addition. (2S,3R,4S,5S,6S)-2-(((2-(2-(2-Azidoethoxy)ethoxy)-4- (hydroxymethyl)phenyl)carbamoyl)oxy)-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (136) can be prepared by reacting 2S,3R,4S,5S,6S)-2-(((2-(2-(2-azidoethoxy)ethoxy)-4- (((tert-butyldimethylsilyl)oxy)methyl)phenyl)carbamoyl)oxy)-6-(methoxycarbonyl)tetrahydro-2H- pyran-3,4,5-triyl triacetate (135) with p-toluenesulfonic acid monohydrate. The reaction is typically performed at ambient temperature in a solvent such as, but not limited to methanol.
(2S,3R,4S,5S,6S)-2-(((2-(2-(2-Azidoethoxy)ethoxy)-4-(hydroxymethyl)phenyl)carbamoyl)oxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (136) can be reacted with bis(4- nitrophenyl)carbonate in the presence of a base such as, but not limited to, N,N- diisopropylethylamine, to provide (2S,3R,4S,5S,6S)-2-(((2-(2-(2-azidoethoxy)ethoxy)-4-((((4- nitrophenoxy)carbonyl)oxy)methyl)phenyl)carbamoyl)oxy)-6-(methoxycarbonyl)tetrahydro-2H- pyran-3,4,5-triyl triacetate (137). The reaction is typically performed at ambient temperature in a solvent such as, but not limited to, N,N-dimethylformamide. (2S,3R,4S,5S,6S)-2-(((2-(2-(2- Azidoethoxy)ethoxy)-4-((((4-nitrophenoxy)carbonyl)oxy)methyl)phenyl)carbamoyl)oxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (137) can be reacted with compound in the presence of a base such as, but not limited to, N,N-diisopropylethylamine, followed by treatment with aqueous lithium hydroxide, to provide compound (138). The first step is typically conducted at ambient temperature in a solvent such as, but not limited to N,N-dimethylformamide, and the second step is typically conducted at low temperature in a solvent such as but not limited to methanol.
Compound (138) can be treated with tris(2-carboxyethyl))phosphine hydrochloride, followed by reaction with compound (84) in the presence of a base such as, but not limited to, N,N- diisopropylethylamine, to provide compound (139). The reaction with tris(2-carboxyethyl))phosphine hydrochloride is typically performed at ambient temperature in a solvent such as, but not limited to, tetrahydrofuran, water, or mixtures thereof, and the reaction with N-succinimidyl 6- maleimidohexanoate is typically performed at ambient temperature in a solvent such as, but not limited to, N,N-dimethylformamide.
.1 v
Figure imgf000251_0001
4 4
92 99 24 E1 Scheme 20 describes the synthesis of galactoside linker intermediates and synthons.
(2S,3R,4S,5S,6R)-6-(Acetoxymethyl)tetrahydro-2H-pyran-2,3,4,5-tetrayl tetraacetate (140) can be treated with HBr in acetic acid to provide (2R,3S,4S,5R,6S)-2-(acetoxymethyl)-6-bromotetrahydro- 2H-pyran-3,4,5-triyl triacetate (141). The reaction is typically performed at ambient temperature under a nitrogen atmosphere. (2R,3S,4S,5R,6S)-2-(Acetoxymethyl)-6-(4-formyl-2- nitrophenoxy)tetrahydro-2H-pyran-3,4,5-triyl triacetate (143) can be prepared by treating
(2R,3S,4S,5R,6S)-2-(acetoxymethyl)-6-bromotetrahydro-2H-pyran-3,4,5-triyl triacetate (141) with silver(I) oxide in the presence of 4-hydroxy-3-nitrobenzaldehyde (142). The reaction is typically performed at ambient temperature in a solvent such as, but not limited to, acetonitrile.
(2R,3S,4S,5R,6S)-2-(Acetoxymethyl)-6-(4-formyl-2-nitrophenoxy)tetrahydro-2H-pyran-3,4,5-triyl triacetate (143) can be treated with sodium borohydride to provide (2R,3S,4S,5R,6S)-2- (acetoxymethyl)-6-(4-(hydroxymethyl)-2-nitrophenoxy)tetrahydro-2H-pyran-3,4,5-triyl triacetate (144). The reaction is typically performed at low temperature in a solvent such as but not limited to tetrahydrofuran, methanol, or mixtures thereof. (2R,3S,4S,5R,6S)-2-(Acetoxymethyl)-6-(2-amino-4- (hydroxymethyl)phenoxy)tetrahydro-2H-pyran-3,4,5-triyl triacetate (145) can be prepared by treating (2R,3S,4S,5R,6S)-2-(acetoxymethyl)-6-(4-(hydroxymethyl)-2-nitrophenoxy)tetrahydro-2H-pyran- 3,4,5-triyl triacetate (144) with zinc in the presence of hydrochloric acid. The reaction is typically performed at low temperature, under a nitrogen atmosphere, in a solvent such as, but not limited to, tetrahydrofuran. (2S,3R,4S,5S,6R)-2-(2-(3-((((9H-Fluoren-9- yl)methoxy)carbonyl)amino)propanamido)-4-(hydroxymethyl)phenoxy)-6- (acetoxymethyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (146) can be prepared by reacting
(2R,3S,4S,5R,6S)-2-(acetoxymethyl)-6-(2-amino-4-(hydroxymethyl)phenoxy)tetrahydro-2H-pyran- 3,4,5-triyl triacetate (145) with (9H-fluoren-9-yl)methyl (3-chloro-3-oxopropyl)carbamate (103) in the presence of a base such as, but not limited to, N,N-diisopropylethylamine. The reaction is typically performed at low temperature, in a solvent such as, but not limited to, dichloromethane. (2S,3R,4S,5S,6R)-2-(2-(3-((((9H-Fluoren-9-yl)methoxy)carbonyl)amino)propanamido)-4- (hydroxymethyl)phenoxy)-6-(acetoxymethyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (146) can be reacted with bis(4-nitrophenyl)carbonate in the presence of a base such as, but not limited to, N,N- diisopropylethylamine, to provide (2S,3R,4S,5S,6R)-2-(2-(3-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)propanamido)-4-((((4-nitrophenoxy)carbonyl)oxy)methyl)phenoxy)-6- (acetoxymethyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (147). The reaction is typically performed at low temperature, in a solvent such as, but not limited to, N,N-dimethylformamide.
(2S,3R,4S,5S,6R)-2-(2-(3-((((9H-Fluoren-9-yl)methoxy)carbonyl)amino)propanamido)-4-((((4- nitrophenoxy)carbonyl)oxy)methyl)phenoxy)-6-(acetoxymethyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (147) can be reacted with compound (88) in the presence of a base such as, but not limited to N,N-diisopropylethylamine, followed by treatment with lithium hydroxide, to provide compound (148) The first step is typically performed at low temperature in a solvent such as but not limited to N,N-dimethylformamide, and the second step is typically performed at ambient temperature, in a solvent such as, but not limited to, methanol. Compound (148) can be treated with compound (84), wherein Sp is a spacer, in the presence of a base, such as, but not limited to N,N- diisopropylethylamine, to provide compound (149). The reaction is typically performed at ambient temperature, in a solvent such as, but not limited to, N,N-dimethylformamide. III.A.7. General Methods for Synthesizing Anti-B7-H3 ADCs
The present invention also discloses a process to prepare an anti-B7-H3 ADC according to structural formula (I):
Figure imgf000253_0001
wherein D, L, LK, Ab and m are as defined in the Detailed Description section. The process comprises:
treating an antibody in an aqueous solution with an effective amount of a disulfide reducing agent at 30-40 °C for at least 15 minutes, and then cooling the antibody solution to 20-27 °C;
adding to the reduced antibody solution a solution of water/dimethyl sulfoxide comprising a synthon selected from the group of 2.1 to 2.31 and 2.34 to 2.72 (Table B);
adjusting the pH of the solution to a pH of 7.5 to 8.5; and
allowing the reaction to run for 48 to 80 hours to form the ADC;
wherein the mass is shifted by 18 ± 2 amu for each hydrolysis of a succinimide to a succinamide as measured by electron spray mass spectrometry; and
wherein the ADC is optionally purified by hydrophobic interaction chromatography.
In certain embodiments, Ab is an anti-B7-H3 antibody, wherein the hB7-H3 antibody comprises the heavy and light chain CDRs of huAb13v1, huAb3v2.5, or huAb3v2.6;
The present invention is also directed to an anti-B7-H3 ADC prepared by the above-described process.
In one embodiment, the anti-B7-H3 ADC disclosed in the present application is formed by contacting an antibody that binds an B7-H3 cell surface receptor or tumor associated antigen expressed on a tumor cell with a drug-linker synthon under conditions in which the drug-linker synthon covalently links to the antibody through a maleimide moiety as shown in formula (IId) or (IIe),
Figure imgf000253_0002
, wherein D is the Bcl-xL inhibitor drug according to structural formula (IIa) or (IIb) as described above and L1 is the portion of the linker not formed from the maleimide upon attachment of the synthon to the antibody; and wherein the drug-linker synthon is selected from the group consisting of synthon examples 2.1 to2.31 and 2.34 to 2.72 (Table B), or a pharmaceutically acceptable salt thereof.
In certain embodiments, the contacting step is carried out under conditions such that the anti- B7-H3 ADC has a DAR of 1.5, 2, 3 or 4. III.B. Anti-B7-H3 ADCs: Other Exemplary Drugs for Conjugation
Anti-B7-H3 antibodies may be used in ADCs to target one or more drug(s) to a cell of interest, e.g., a cancer cell expressing B7-H3. The anti-B7-H3 ADCs of the invention provide a targeted therapy that may, for example, reduce the side effects often seen with anti-cancer therapies, as the one or more drug(s) is delivered to a specific cell. Auristatins
Anti-B7-H3 antibodies of the invention, e.g., the huAb13v1, huAb3v2.5, or huAb3v2.6 antibody, may be conjugated to at least one auristatin. Auristatins represent a group of dolastatin analogs that have generally been shown to possess anticancer activity by interfering with microtubule dynamics and GTP hydrolysis, thereby inhibiting cellular division. For example, auristatin E (U.S. Patent No.5,635,483) is a synthetic analogue of the marine natural product dolastatin 10, a compound that inhibits tubulin polymerization by binding to the same site on tubulin as the anticancer drug vincristine (G. R. Pettit, Prog. Chem. Org. Nat. Prod, 70: 1-79 (1997)). Dolastatin 10, auristatin PE, and auristatin E are linear peptides having four amino acids, three of which are unique to the dolastatin class of compounds. Exemplary embodiments of the auristatin subclass of mitotic inhibitors include, but are not limited to, monomethyl auristatin D (MMAD or auristatin D derivative), monomethyl auristatin E (MMAE or auristatin E derivative), monomethyl auristatin F (MMAF or auristatin F derivative), auristatin F phenylenediamine (AFP), auristatin EB (AEB), auristatin EFP (AEFP), and 5-benzoylvaleric acid-AE ester (AEVB). The synthesis and structure of auristatin derivatives are described in U.S. Patent Application Publication Nos.2003-0083263, 2005- 0238649 and 2005-0009751; International Patent Publication No. WO 04/010957, International Patent Publication No. WO 02/088172, and U.S. Pat. Nos.6,323,315; 6,239,104; 6,034,065; 5,780,588; 5,665,860; 5,663,149; 5,635,483; 5,599,902; 5,554,725; 5,530,097; 5,521,284; 5,504,191; 5,410,024; 5,138,036; 5,076,973; 4,986,988; 4,978,744; 4,879,278; 4,816,444; and 4,486,414, each of which is incorporated by reference herein.
In one embodiment, anti-B7-H3 antibodies of the invention, e.g., huAb13v1, huAb3v2.5, or huAb3v2.6, are conjugated to at least one MMAE (mono-methyl auristatin E). Monomethyl auristatin E (MMAE, vedotin) inhibits cell division by blocking the polymerization of tubulin. However, due to its super toxicity, auristatin E cannot be used as a drug itself. Auristatin E can be linked to a monoclonal antibody (mAb) that recognizes a specific marker expression in cancer cells and directs MMAE to the cancer cells. In one embodiment, the linker linking MMAE to the anti-B7-H3 antibody is stable in extracellular fluid (i.e., the medium or environment that is external to cells), but is cleaved by cathepsin once the ADC has bound to the specific cancer cell antigen and entered the cancer cell, thus releasing the toxic MMAE and activating the potent anti-mitotic mechanism.
In one embodiment, an anti-B7-H3 antibody described herein, e.g., huAb13v1, huAb3v2.5, or huAb3v2.6, is conjugated to at least one MMAF (monomethylauristatin F). Monomethyl auristatin F (MMAF) inhibits cell division by blocking the polymerization of tubulin. It has a charged C-terminal phenylalanine residue that attenuates its cytotoxic activity compared to its uncharged counterpart MMAE. However, due to its super toxicity, auristatin F cannot be used as a drug itself, but can be linked to a monoclonal antibody (mAb) that directs it to the cancer cells. In one embodiment, the linker to the anti-B7-H3 antibody is stable in extracellular fluid, but is cleaved by cathepsin once the conjugate has entered a tumor cell, thus activating the anti-mitotic mechanism.
The
Figure imgf000255_0001
An example of huAb13v1, huAb3v2.5, or huAb3v2.6-vcMMAE is also provided in Figure 3. Notably, Figure 3 describes a situation where the antibody (e.g., huAb13v1, huAb3v2.5, or huAb3v2.6) is coupled to a single drug and, therefore, has a DAR of 1. In certain embodiments, the ADC will have a DAR of 2 to 8, or, alternatively, 2 to 4. Other Drugs for Conjugation
Examples of drugs that may be used in ADCs, i.e., drugs that may be conjugated to the anti- B7-H3 antibodies of the invention, are provided below, and include mitotic inhibitors, antitumor antibiotics, immunomodulating agents, gene therapy vectors, alkylating agents, antiangiogenic agents, antimetabolites, boron-containing agents, chemoprotective agents, hormone agents, glucocorticoids, photoactive therapeutic agents, oligonucleotides, radioactive isotopes, radiosensitizers, topoisomerase inhibitors, kinase inhibitors, and combinations thereof. 1. Mitotic Inhibitors
In one aspect, anti-B7-H3 antibodies may be conjugated to one or more mitotic inhibitor(s) to form an ADC for the treatment of cancer. The term“mitotic inhibitor”, as used herein, refers to a cytotoxic and/or therapeutic agent that blocks mitosis or cell division, a biological process particularly important to cancer cells. A mitotic inhibitor disrupts microtubules such that cell division is prevented, often by effecting microtubule polymerization (e.g., inhibiting microtubule
polymerization) or microtubule depolymerization (e.g., stabilizing the microtubule cytoskeleton against depolymrization). Thus, in one embodiment, an anti-B7-H3 antibody of the invention is conjugated to one or more mitotic inhibitor(s) that disrupts microtubule formation by inhibiting tubulin polymerization. In another embodiment, an anti-B7-H3 antibody of the invention is conjugated to one or more mitotic inhibitor(s) that stabilizes the microtubule cytoskeleton from deploymerization. In one embodiment, the mitotic inhibitor used in the ADCs of the invention is Ixempra (ixabepilone). Examples of mitotic inhibitors that may be used in the anti-B7-H3 ADCs of the invention are provided below. Included in the genus of mitotic inhibitors are auristatins, described above. a. Dolastatins
The anti-B7-H3 antibodies of the invention may be conjugated to at least one dolastatin to form an ADC. Dolastatins are short peptidic compounds isolated from the Indian Ocean sea hare Dolabella auricularia (see Pettit et al., J. Am. Chem. Soc., 1976, 98, 4677). Examples of dolastatins include dolastatin 10 and dolatstin 15. Dolastatin 15, a seven-subunit depsipeptide derived from Dolabella auricularia, and is a potent antimitotic agent structurally related to the antitubulin agent dolastatin 10, a five-subunit peptide obtained from the same organism. Thus, in one embodiment, the anti-B7-H3 ADC of the invention comprises an anti-B7-H3 antibody, as described herein, and at least one dolastatin. Auristatins, described above, are synthetic derivatives of dolastatin 10. b. Maytansinoids
The anti-B7-H3 antibodies of the invention may be conjugated to at least one maytansinoid to form an ADC. Maytansinoids are potent antitumor agents that were originally isolated from members of the higher plant families Celastraceae, Rhamnaceae, and Euphorbiaceae, as well as some species of mosses (Kupchan et al, J. Am. Chem. Soc.94:1354-1356 [1972]; Wani et al, J. Chem. Soc. Chem. Commun.390: [1973]; Powell et al, J. Nat. Prod.46:660-666 [1983]; Sakai et al, J. Nat. Prod.51 :845-850 [1988]; and Suwanborirux et al, Experientia 46:117-120 [1990]). Evidence suggests that maytansinoids inhibit mitosis by inhibiting polymerization of the microtubule protein tubulin, thereby preventing formation of microtubules (see, e.g., U.S. Pat. No.6,441,163 and Remillard et al., Science, 189, 1002-1005 (1975)). Maytansinoids have been shown to inhibit tumor cell growth in vitro using cell culture models, and in vivo using laboratory animal systems. Moreover, the cytotoxicity of maytansinoids is 1,000-fold greater than conventional chemotherapeutic agents, such as, for example, methotrexate, daunorubicin, and vincristine (see, e.g., U.S. Pat. No.5,208,020).
Maytansinoids to include maytansine, maytansinol, C-3 esters of maytansinol, and other maytansinol analogues and derivatives (see, e.g., U.S. Pat. Nos.5,208,020 and 6,441,163, each of which is incorporated by reference herein). C-3 esters of maytansinol can be naturally occurring or synthetically derived. Moreover, both naturally occurring and synthetic C-3 maytansinol esters can be classified as a C-3 ester with simple carboxylic acids, or a C-3 ester with derivatives of N-methyl-L- alanine, the latter being more cytotoxic than the former. Synthetic maytansinoid analogues are described in, for example, Kupchan et al., J. Med. Chem., 21, 31-37 (1978).
Suitable maytansinoids for use in ADCs of the invention can be isolated from natural sources, synthetically produced, or semi-synthetically produced. Moreover, the maytansinoid can be modified in any suitable manner, so long as sufficient cytotoxicity is preserved in the ultimate conjugate molecule. In this regard, maytansinoids lack suitable functional groups to which antibodies can be linked. A linking moiety desirably is utilized to link the maytansinoid to the antibody to form the conjugate, and is described in more detail in the linker section below. The structure of an exemplary maytansinoid, mertansine (DM1), is provided below.
Representative examples of maytansinoids include, but are not limited, to DM1 (N2'-deacetyl- N2'-(3-mercapto-1-oxopropyl)-maytansine; also referred to as mertansine, drug maytansinoid 1; ImmunoGen, Inc.; see also Chari et al. (1992) Cancer Res 52:127), DM2, DM3 (N2'-deacetyl-N2'-(4- mercapto-1-oxopentyl)-maytansine), DM4 (4-methyl-4-mercapto-1-oxopentyl)-maytansine), and maytansinol (a synthetic maytansinoid analog). Other examples of maytansinoids are described in US Patent No.8,142,784, incorporated by reference herein.
Ansamitocins are a group of maytansinoid antibiotics that have been isolated from various bacterial sources. These compounds have potent antitumor activities. Representative examples include, but are not limited to ansamitocin P1, ansamitocin P2, ansamitocin P3, and ansamitocin P4.
In one embodiment of the invention, an anti-B7-H3 antibody is conjugated to at least one DM1. In one embodiment, an anti-B7-H3 antibody is conjugated to at least one DM2. In one embodiment, an anti-B7-H3 antibody is conjugated to at least one DM3. In one embodiment, an anti- B7-H3 antibody is conjugated to at least one DM4. d. Plant Alkaloids
The anti-B7-H3 antibodies of the invention may be conjugated to at least one plant alkaloid, e.g., a taxane or vinca alkaloid. Plant alkaloids are chemotherapy treatments derived made from certain types of plants. The vinca alkaloids are made from the periwinkle plant (catharanthus rosea), whereas the taxanes are made from the bark of the Pacific Yew tree (taxus). Both the vinca alkaloids and taxanes are also known as antimicrotubule agents, and are described in more detail below. Taxanes
Anti-B7-H3 antibodies described herein may be conjugated to at least one taxane. The term “taxane” as used herein refers to the class of antineoplastic agents having a mechanism of microtubule action and having a structure that includes the taxane ring structure and a stereospecific side chain that is required for cytostatic activity. Also included within the term“taxane” are a variety of known derivatives, including both hydrophilic derivatives, and hydrophobic derivatives. Taxane derivatives include, but not limited to, galactose and mannose derivatives described in International Patent Application No. WO 99/18113; piperazino and other derivatives described in WO 99/14209; taxane derivatives described in WO 99/09021, WO 98/22451, and U.S. Pat. No.5,869,680; 6-thio derivatives described in WO 98/28288; sulfenamide derivatives described in U.S. Pat. No.5,821,263; and taxol derivative described in U.S. Pat. No.5,415,869, each of which is incorporated by reference herein. Taxane compounds have also previously been described in U.S. Pat. Nos.5,641,803, 5,665,671, 5,380,751, 5,728,687, 5,415,869, 5,407,683, 5,399,363, 5,424,073, 5,157,049, 5,773,464, 5,821,263, 5,840,929, 4,814,470, 5,438,072, 5,403,858, 4,960,790, 5,433,364, 4,942,184, 5,362,831, 5,705,503, and 5,278,324, all of which are expressly incorporated by reference. Further examples of taxanes include, but are not limited to, docetaxel (Taxotere; Sanofi Aventis), paclitaxel (Abraxane or Taxol; Abraxis Oncology), carbazitaxel, tesetaxel, opaxio, larotaxel, taxoprexin, BMS-184476, hongdoushan A, hongdoushan B, and hongdoushan C, and nanoparticle paclitaxel (ABI-007 / Abraxene; Abraxis Bioscience).
In one embodiment, the anti-B7-H3 antibody of the invention is conjugated to at least one docetaxel molecule. In one embodiment, the anti-B7-H3 antibody of the invention is conjugated to at least one paclitaxel molecule. Vinca alkaloids
In one embodiment, the anti-B7-H3 antibody is conjugated to at least one vinca alkaloid. Vinca alkaloids are a class of cell-cycle-specific drugs that work by inhibiting the ability of cancer cells to divide by acting upon tubulin and preventing the formation of microtubules. Examples of vinca alkaloids that may be used in the ADCs of the invention include, but are not limited to, vindesine sulfate, vincristine, vinblastine, and vinorelbine. 2. Antitumor Antibiotics
Anti-B7-H3 antibodies of the invention may be conjugated to one or more antitumor antibiotic(s) for the treatment of cancer. As used herein, the term“antitumor antibiotic” means an antineoplastic drug that blocks cell growth by interfering with DNA and is made from a
microorganism. Often, antitumor antibiotics either break up DNA strands or slow down or stop DNA synthesis. Examples of antitumor antibiotics that may be included in the anti-B7-H3 ADCs of the invention include, but are not limited to, actinomycines (e.g., pyrrolo[2,1-c][1,4]benzodiazepines), anthracyclines, calicheamicins, and duocarmycins, described in more detail below. a. Actinomycins
The anti-B7-H3 antibodies of the invention may be conjugated to at least one actinomycin. Actinomycins are a subclass of antitumor antibiotics isolated from bacteria of the genus Streptomyces. Representative examples actinomycins include, but are not limited to, actinomycin D (Cosmegen [also known as actinomycin, dactinomycin, actinomycin IV, actinomycin C1], Lundbeck, Inc.), anthramycin, chicamycin A, DC-81, mazethramycin, neothramycin A, neothramycin B,
porothramycin, prothracarcin B, SG2285, sibanomicin, sibiromycin, and tomaymycin. In one embodiment, the anti-B7-H3 antibody of the invention is conjugated to at least one
pyrrolobenzodiazepine (PBD). Examples of PBDs include, but are not limited to, anthramycin, chicamycin A, DC-81, mazethramycin, neothramycin A, neothramycin B, porothramycin, prothracarcin B, SG2000 (SJG-136), SG2202 (ZC-207), SG2285 (ZC-423), sibanomicin, sibiromycin and tomaymycin. Thus, in one embodiment, anti-B7-H3 antibodies of the invention are conjugated to at least one actinomycin, e.g., actinomycin D, or at least one PBD, e.g., a pyrrolobenzodiazepine (PBD) dimer.
The structures of PBDs can be found, for example, in U.S. Patent Application Pub. Nos.
2013/0028917 and 2013/0028919, and in WO 2011/130598 A1, each of which are incorporated herein by reference in their entirety. The generic structure of a PBD is provided below.
Figure imgf000260_0001
PBDs differ in the number, type and position of substituents, in both their aromatic A rings and pyrrolo C rings, and in the degree of saturation of the C ring. In the B-ring, there is generally an imine (N=C), a carbinolamine (NH-CH(OH)), or a carbinolamine methyl ether (NH-CH(OMe)) at the N10-C11 position which is the electrophilic centre responsible for alkylating DNA. All of the known natural products have an (S)-configuration at the chiral C11α position which provides them with a right-handed twist when viewed from the C ring towards the A ring. The PBD examples provided herein may be conjugated to the anti-B7-H3 antibodies of the invention. Further examples of PBDs which may be conjugated to the anti-B7-H3 antibodies of the invention can be found, for example, in U.S. Patent Application Publication Nos.2013/0028917 A1 and 2013/0028919 A1, in U.S. Patent Nos.7,741,319 B2 , and in WO 2011/130598 A1 and WO 2006/111759 A1, each of which are incorporated herein by reference in their entirety.
A representative PBD dimer having the following formula XXX may be conjugated to the anti-B7-H3 antibodies of the invention:
Figure imgf000261_0001
(XXX)
wherein:
R30 is of formula XXXI:
Figure imgf000261_0002
where A is a C5-7 aryl group, X is a group conjugated to the Linker unit selected from the group consisting of—O—,—S—,—C(O)O—,—C(O)—,—NH(C═O)—, and—N(RN)—, wherein RN is selected from the group consisting of H, C1-4 alkyl and (C2H4O)mCH3, where s is 1 to 3, and either:
(i) Q1 is a single bond, and Q2 is selected from the group consisting of a single bond and—Z— (CH2)n—, where Z is selected from the group consisting of a single bond, O, S and NH and n is from 1 to 3; or
(ii) Q1 is—CH═CH—, and Q2 is a single bond;
R130 is a C5-10 aryl group, optionally substituted by one or more substituents selected from the group consisting of halo, nitro, cyano, C1-12 alkoxy, C3-20 heterocycloalkoxy, C5-20 aryloxy, heteroaryloxy, alkylalkoxy, arylalkoxy, alkylaryloxy, heteroarylalkoxy, alkylheteroaryloxy, C1-7 alkyl, C3-7 heterocyclyl and bis-oxy-C1-3 alkylene;
R31 and R33 are independently selected from the group consisting of H, Rx, OH, ORx, SH, SRx, NH2, NHRx, NRxRxx′, nitro, Me3Sn and halo;
where R and R′ are independently selected from the group consisting of optionally substituted C1-12 alkyl, C3-20 heterocyclyl and C5-20 aryl groups;
R32 is selected from the group consisting of H, Rx, OH, ORx, SH, SRx, NH2, NHRx, NHRxRxx, nitro, Me3Sn and halo; either:
(a) R34 is H, and R11 is OH, ORxA, where RxA is C1-4 alkyl;
(b) R34 and R35 form a nitrogen-carbon double bond between the nitrogen and carbon atoms to which they are bound; or
(c) R34is H and R35 is SOzM, where z is 2 or 3;
Rxxx is a C3-12 alkylene group, which chain may be interrupted by one or more heteroatoms, selected from the group consisting of O, S, NH, and an aromatic ring;
Yx and Yx′ are is selected from the group consisting of O, S, and NH;
R31′, R32′, R33′ are selected from the same groups as R31, R32 and R33 respectively and R34′ and R35′ are the same as R34 and R35, and each M is a monovalent pharmaceutically acceptable cation or both M groups together are a divalent pharmaceutically acceptable cation.
C1-12 alkyl: The term“C1-12 alkyl” as used herein, pertains to a monovalent moiety obtained by removing a hydrogen atom from a carbon atom of a hydrocarbon compound having from 1 to 12 carbon atoms, which may be aliphatic or alicyclic, and which may be saturated or unsaturated (e.g. partially unsaturated, fully unsaturated). Thus, the term“alkyl” includes the sub-classes alkenyl, alkynyl, cycloalkyl, etc., discussed below.
Examples of saturated alkyl groups include, but are not limited to, methyl (C1), ethyl (C2), propyl (C3), butyl (C4), pentyl (C5), hexyl (C6) and heptyl (C7).
Examples of saturated linear alkyl groups include, but are not limited to, methyl (C1), ethyl (C2), n-propyl (C3), n-butyl (C4), n-pentyl (amyl) (C5), n-hexyl (C6) and n-heptyl (C7).
Examples of saturated branched alkyl groups include iso-propyl (C3), iso-butyl (C4), sec-butyl (C4), tert-butyl (C4), iso-pentyl (C5), and neo-pentyl (C5).
C3-20 heterocyclyl: The term“C3-20 heterocyclyl” as used herein, pertains to a monovalent moiety obtained by removing a hydrogen atom from a ring atom of a heterocyclic compound, which moiety has from 3 to 20 ring atoms, of which from 1 to 10 are ring heteroatoms. Preferably, each ring has from 3 to 7 ring atoms, of which from 1 to 4 are ring heteroatoms.
In this context, the prefixes (e.g. C3-20, C3-7, C5-6, etc.) denote the number of ring atoms, or range of number of ring atoms, whether carbon atoms or heteroatoms. For example, the term“C5- 6heterocyclyl”, as used herein, pertains to a heterocyclyl group having 5 or 6 ring atoms.
Examples of monocyclic heterocyclyl groups include, but are not limited to, those derived from:
N1: aziridine (C3), azetidine (C4), pyrrolidine (tetrahydropyrrole) (C5), pyrroline (e.g., 3- pyrroline, 2,5-dihydropyrrole) (C5), 2H-pyrrole or 3H-pyrrole (isopyrrole, isoazole) (C5), piperidine (C6), dihydropyridine (C6), tetrahydropyridine (C6), azepine (C7); O1: oxirane (C3), oxetane (C4), oxolane (tetrahydrofuran) (C5), oxole (dihydrofuran) (C5), oxane (tetrahydropyran) (C6), dihydropyran (C6), pyran (C6), oxepin (C7); S1: thiirane (C3), thietane (C4), thiolane (tetrahydrothiophene) (C5), thiane (tetrahydrothiopyran) (C6) thiepane (C7); O2: dioxolane (C5) dioxane (C6) and dioxepane (C7); O3: trioxane (C6); N2: imidazolidine (C5), pyrazolidine (diazolidine) (C5), imidazoline (C5), pyrazoline (dihydropyrazole) (C5), piperazine (C6); N1O1: tetrahydrooxazole (C5), dihydrooxazole (C5), tetrahydroisoxazole (C5), dihydroisoxazole (C5), morpholine (C6), tetrahydrooxazine (C6), dihydrooxazine (C6), oxazine (C6); N1S1: thiazoline (C5), thiazolidine (C5), thiomorpholine (C6); N2O1: oxadiazine (C6); O1S1: oxathiole (C5) and oxathiane (thioxane) (C6); and, N1O1S1: oxathiazine (C6).
Examples of substituted monocyclic heterocyclyl groups include those derived from saccharides, in cyclic form, for example, furanoses (C5), such as arabinofuranose, lyxofuranose, ribofuranose, and xylofuranse, and pyranoses (C6), such as allopyranose, altropyranose,
glucopyranose, mannopyranose, gulopyranose, idopyranose, galactopyranose, and talopyranose.
C5-20 aryl: The term“C5-20 aryl”, as used herein, pertains to a monovalent moiety obtained by removing a hydrogen atom from an aromatic ring atom of an aromatic compound, which moiety has from 3 to 20 ring atoms. Preferably, each ring has from 5 to 7 ring atoms.
In this context, the prefixes (e.g. C3-20, C5-7, C5-6, etc.) denote the number of ring atoms, or range of number of ring atoms, whether carbon atoms or heteroatoms. For example, the term "C5-6 aryl" as used herein, pertains to an aryl group having 5 or 6 ring atoms.
In one embodiment, the anti-B7-H3 antibodies of the invention may be conjugated to a PBD dimer having the following formula XXXIa:
Figure imgf000263_0001
(XXXIa) wherein the above structure describes the PBD dimer SG2202 (ZC-207) and is conjugated to the anti- B7-H3 antibody of the invention via a linker L. SG2202 (ZC-207) is disclosed in, for example, U.S. Patent App. Pub. No.2007/0173497, which is incorporated herein by reference in its entirety.
In another embodiment, a PBD dimer, SGD-1882, is conjugated to anti-B7-H3 antibody of the invention via a drug linker, as depicted in Figure 4. SGD-1882 is disclosed in Sutherland et al. (2013) Blood 122(8):1455 and in U.S Patent App. Pub. No.2013/0028919, which is incorporated herein be reference in its entirety. As described in Figure 4, the PBD dimer SGD-1882 may be conjugated to an antibody via an mc-val-ala-dipeptide linker (collectively referred to as SGD-1910 in Figure 4). In a certain embodiment, an anti-B7-H3 antibody, as disclosed herein, is conjugated to the PBD dimer described in Figure 4. Thus, in a further embodiment, the invention includes an anti-B7- H3 antibody, as disclosed herein, conjugated to a PBD dimer via a mc-val-ala-dipeptide linker, as described in Figure 4. In certain embodiments, the invention includes an anti-B7-H3 antibody comprising a heavy chain variable region comprising a CDR3 domain comprising the amino acid sequence of SEQ ID NO: 35, a CDR2 domain comprising the amino acid sequence of SEQ ID NO: 34, and a CDR1 domain comprising the amino acid sequence of SEQ ID NO: 33, and a light chain variable region comprising a CDR3 domain comprising the amino acid sequence of SEQ ID NO: 39, a CDR2 domain comprising the amino acid sequence of SEQ ID NO: 38, and a CDR1 domain comprising the amino acid sequence of SEQ ID NO: 37, conjugated to a PBD, including, but not limited to, the PBD dimer described in Figure 4. In certain embodiments, the invention includes an anti-B7-H3 antibody comprising a heavy chain variable region comprising a CDR3 domain comprising the amino acid sequence of SEQ ID NO: 12, a CDR2 domain comprising the amino acid sequence of SEQ ID NO: 140, and a CDR1 domain comprising the amino acid sequence of SEQ ID NO: 10, and a light chain variable region comprising a CDR3 domain comprising the amino acid sequence of SEQ ID NO: 15, a CDR2 domain comprising the amino acid sequence of SEQ ID NO: 7, and a CDR1 domain comprising the amino acid sequence of SEQ ID NO: 136, conjugated to a PBD, including, but not limited to, the PBD dimer described in Figure 4. In certain embodiments, the invention includes an anti-B7-H3 antibody comprising a heavy chain variable region comprising a CDR3 domain comprising the amino acid sequence of SEQ ID NO: 12, a CDR2 domain comprising the amino acid sequence of SEQ ID NO: 140, and a CDR1 domain comprising the amino acid sequence of SEQ ID NO: 10, and a light chain variable region comprising a CDR3 domain comprising the amino acid sequence of SEQ ID NO: 15, a CDR2 domain comprising the amino acid sequence of SEQ ID NO: 7, and a CDR1 domain comprising the amino acid sequence of SEQ ID NO: 138, conjugated to a PBD, including, but not limited to, the PBD dimer described in Figure 4. In certain embodiments, the invention includes an anti-B7-H3 antibody comprising the heavy chain variable region of huAb13v1 as defined by the amino acid sequence set forth in SEQ ID NO: 147, or huAb3v2.5 or huAb3v2.6 as defined by the amino acid sequence set forth in SEQ ID NO: 139, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 144, 135, or 137 corresponding to huAb13v1, huAb3v2.5, or huAb3v2.6, respectively, wherein the antibody is conjugated to a PBD, such as, but not limited to, the exemplary PBD dimer of Figure 4.
b. Anthracyclines
Anti-B7-H3 antibodies of the invention may be conjugated to at least one anthracycline.
Anthracyclines are a subclass of antitumor antibiotics isolated from bacteria of the genus
Streptomyces. Representative examples include, but are not limited to daunorubicin (Cerubidine, Bedford Laboratories), doxorubicin (Adriamycin, Bedford Laboratories; also referred to as doxorubicin hydrochloride, hydroxydaunorubicin, and Rubex), epirubicin (Ellence, Pfizer), and idarubicin (Idamycin; Pfizer Inc.). Thus, in one embodiment, the anti-B7-H3 antibody of the invention is conjugated to at least one anthracycline, e.g., doxorubicin. c. Calicheamicins
The anti-B7-H3 antibodies of the invention may be conjugated to at least one calicheamicin. Calicheamicins are a family of enediyne antibiotics derived from the soil organism Micromonospora echinospora. Calicheamicins bind the minor groove of DNA and induce double-stranded DNA breaks, resulting in cell death with a 100 fold increase over other chemotherapeutics (Damle et al. (2003) Curr Opin Pharmacol 3:386). Preparation of calicheamicins that may be used as drug conjugates in the invention have been described, see U.S. Pat. Nos.5,712,374; 5,714,586; 5,739,116; 5,767,285; 5,770,701; 5,770,710; 5,773,001; and 5,877,296. Structural analogues of calicheamicin which may be used include, but are not limited to, γ I
1, α I
2, α I
3, N-acetyl-γ I
1 , PSAG and θI
1 (Hinman et al., Cancer Research 53:3336-3342 (1993), Lode et al., Cancer Research 58:2925-2928 (1998) and the aforementioned U.S. Patent Nos.5,712,374; 5,714,586; 5,739,116; 5,767,285; 5,770,701;
5,770,710; 5,773,001; and 5,877,296). Thus, in one embodiment, the anti-B7-H3 antibody of the invention is conjugated to at least one calicheamicin. d. Duocarmycins
Anti-B7-H3 antibodies of the invention may be conjugated to at least one duocarmycin. Duocarmycins are a subclass of antitumor antibiotics isolated from bacteria of the genus
Streptomyces. (see Nagamura and Saito (1998) Chemistry of Heterocyclic Compounds, Vol. 34, No. 12). Duocarmycins bind to the minor groove of DNA and alkylate the nucleobase adenine at the N3 position (Boger (1993) Pure and Appl Chem 65(6):1123; and Boger and Johnson (1995) PNAS USA 92:3642). Synthetic analogs of duocarmycins include, but are not limited to, adozelesin, bizelesin, and carzelesin. Thus, in one embodiment, the anti-B7-H3 antibody of the invention is conjugated to at least one duocarmycin. e. Other antitumor antibiotics
In addition to the foregoing, additional antitumor antibiotics that may be used in the anti-B7- H3 ADCs of the invention include bleomycin (Blenoxane, Bristol-Myers Squibb), mitomycin, and plicamycin (also known as mithramycin). 3. Immunomodulating Agents
In one aspect, anti-B7-H3 antibodies of the invention may be conjugated to at least one immunomodulating agent. As used herein, the term“immunomodulating agent” refers to an agent that can stimulate or modify an immune response. In one embodiment, an immunomodulating agent is an immunostimulator that enhances a subject’s immune response. In another embodiment, an immunomodulating agent is an immunosuppressant that prevents or decreases a subject’s immune response. An immunomodulating agent may modulate myeloid cells (monocytes, macrophages, dendritic cells, megakaryocytes and granulocytes) or lymphoid cells (T cells, B cells and natural killer (NK) cells) and any further differentiated cell thereof. Representative examples include, but are not limited to, bacillus calmette-guerin (BCG) and levamisole (Ergamisol). Other examples of immunomodulating agents that may be used in the ADCs of the invention include, but are not limited to, cancer vaccines, cytokines, and immunomodulating gene therapy. a. Cancer vaccines
Anti-B7-H3 antibodies of the invention may be conjugated to a cancer vaccine. As used herein, the term“cancer vaccine” refers to a composition (e.g., a tumor antigen and a cytokine) that elicits a tumor-specific immune response. The response is elicited from the subject's own immune system by administering the cancer vaccine, or, in the case of the instant invention, administering an ADC comprising an anti-B7-H3 antibody and a cancer vaccine. In preferred embodiments, the immune response results in the eradication of tumor cells in the body (e.g., primary or metastatic tumor cells). The use of cancer vaccines generally involves the administration of a particular antigen or group of antigens that are, for example, present on the surface a particular cancer cell, or present on the surface of a particular infectious agent shown to facilitate cancer formation. In some
embodiments, the use of cancer vaccines is for prophylactic purposes, while in other embodiments, the use is for therapeutic purposes. Non-limiting examples of cancer vaccines that may be used in the anti-B7-H3 ADCs of the invention include, recombinant bivalent human papillomavirus (HPV) vaccine types 16 and 18 vaccine (Cervarix, GlaxoSmithKline), recombinant quadrivalent human papillomavirus (HPV) types 6, 11, 16, and 18 vaccine (Gardasil, Merck & Company), and sipuleucel- T (Provenge, Dendreon). Thus, in one embodiment, the anti-B7-H3 antibody of the invention is conjugated to at least one cancer vaccine that is either an immunostimulator or is an
immunosuppressant. b. Cytokines
The anti-B7-H3 antibodies of the invention may be conjugated to at least one cytokine. The term“cytokine” generally refers to proteins released by one cell population which act on another cell as intercellular mediators. Cytokines directly stimulate immune effector cells and stromal cells at the tumor site and enhance tumor cell recognition by cytotoxic effector cells (Lee and Margolin (2011) Cancers 3:3856). Numerous animal tumor model studies have demonstrated that cytokines have broad anti-tumor activity and this has been translated into a number of cytokine-based approaches for cancer therapy (Lee and Margoli, supra). Recent years have seen a number of cytokines, including GM-CSF, IL-7, IL-12, IL-15, IL-18 and IL-21, enter clinical trials for patients with advanced cancer (Lee and Margoli, supra). Examples of cytokines that may be used in the ADCs of the invention include, but are not limited to, parathyroid hormone; thyroxine; insulin; proinsulin; relaxin; prorelaxin; glycoprotein hormones such as follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), and luteinizing hormone (LH); hepatic growth factor; fibroblast growth factor; prolactin; placental lactogen; tumor necrosis factor; mullerian-inhibiting substance; mouse gonadotropin-associated peptide; inhibin; activin; vascular endothelial growth factor; integrin; thrombopoietin (TPO); nerve growth factors such as NGF; platelet-growth factor; transforming growth factors (TGFs); insulin-like growth factor-I and -II; erythropoietin (EPO); osteoinductive factors; interferons such as interferon α, γ, colony stimulating factors (CSFs); granulocyte-macrophage-C-SF (GM-CSF); and granulocyte-CSF (G-CSF); interleukins (ILs) such as IL-1, IL-1α, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-11, IL-12; tumor necrosis factor; and other polypeptide factors including LIF and kit ligand (KL). As used herein, the term cytokine includes proteins from natural sources or from recombinant cell culture and biologically active equivalents of the native sequence cytokines. Thus, in one embodiment, the invention provides an ADC comprising an anti-B7-H3 antibody described herein and a cytokine. c. Colony-stimulating factors (CSFs)
The anti-B7-H3 antibodies of the invention may be conjugated to at least one colony stimulating factor (CSF). Colony stimulating factors (CSFs) are growth factors that assist the bone marrow in making white blood cells. Some cancer treatments (e.g., chemotherapy) can affect white blood cells (which help fight infection); therefore, colony-stimulating factors may be introduced to help support white blood cell levels and strengthen the immune system. Colony-stimulating factors may also be used following a bone marrow transplant to help the new marrow start producing white blood cells. Representative examples of CSFs that may be used in the anti-B7-H3 ADCs of the invention include, but are not limited to erythropoietin (Epoetin), filgrastim (Neopogen (also known as granulocyte colony-stimulating factor (G-CSF); Amgen, Inc.), sargramostim (leukine (granulocyte- macrophage colony-stimulating factor and GM-CSF); Genzyme Corporation), promegapoietin, and Oprelvekin (recombinant IL-11; Pfizer, Inc.). Thus, in one embodiment, the invention provides an ADC comprising an anti-B7-H3 antibody described herein and a CSF. 4. Gene Therapy
The anti-B7-H3 antibody of the invention may be conjugated to at least one nucleic acid (directly or indirectly via a carrier) for gene therapy. Gene therapy generally refers to the introduction of genetic material into a cell whereby the genetic material is designed to treat a disease. As it pertains to immunomodulatory agents, gene therapy is used to stimulate a subject's natural ability to inhibit cancer cell proliferation or kill cancer cells. In one embodiment, the anti-B7-H3 ADC of the invention comprises a nucleic acid encoding a functional, therapeutic gene that is used to replace a mutated or otherwise dysfunctional (e.g. truncated) gene associated with cancer. In other
embodiments, the anti-B7-H3 ADC of the invention comprises a nucleic acid that encodes for or otherwise provides for the production of a therapeutic protein to treat cancer. The nucleic acid that encodes the therapeutic gene may be directly conjugated to the anti-B7-H3 antibody, or alternatively, may be conjugated to the anti-B7-H3 antibody through a carrier. Examples of carriers that may be used to deliver a nucleic acid for gene therapy include, but are not limited to, viral vectors or liposomes. 5. Alkylating Agents
The anti-B7-H3 antibodies of the invention may be conjugated to one or more alkylating agent(s). Alkylating agents are a class of antineoplastic compounds that attaches an alkyl group to DNA. Examples of alkylating agents that may be used in the ADCs of the invention include, but are not limited to, alkyl sulfonates, ethylenimimes, methylamine derivatives, epoxides, nitrogen mustards, nitrosoureas, triazines, and hydrazines. a. Alkyl Sulfonates
The anti-B7-H3 antibodies of the invention may be conjugated to at least one alkyl sulfonate. Alkyl sulfonates are a subclass of alkylating agents with a general formula: R-SO2-O-R1, wherein R and R1 are typically alkyl or aryl groups. A representative example of an alkyl sulfonate includes, but is not limited to, busulfan (Myleran, GlaxoSmithKline; Busulfex IV, PDL BioPharma, Inc.). b. Nitrogen Mustards
The anti-B7-H3 antibodies of the invention may be conjugated to at least one nitrogen mustard. Representative examples of this subclass of anti-cancer compounds include, but are not limited to chlorambucil (Leukeran, GlaxoSmithKline), cyclophosphamide (Cytoxan, Bristol-Myers Squibb; Neosar, Pfizer, Inc.), estramustine (estramustine phosphate sodium or Estracyt), Pfizer, Inc.), ifosfamide (Ifex, Bristol-Myers Squibb), mechlorethamine (Mustargen, Lundbeck Inc.), and melphalan (Alkeran or L-Pam or phenylalanine mustard; GlaxoSmithKline). c. Nitrosoureas
The anti-B7-H3 antibody of the invention may be conjugated to at least one nitrosourea. Nitrosoureas are a subclass of alkylating agents that are lipid soluble. Representative examples include, but are not limited to, carmustine (BCNU [also known as BiCNU, N,N-Bis(2-chloroethyl)-N- nitrosourea, or 1, 3-bis (2-chloroethyl)-l-nitrosourea], Bristol-Myers Squibb), fotemustine (also known as Muphoran), lomustine (CCNU or 1-(2-chloro-ethyl)-3-cyclohexyl-1-nitrosourea, Bristol- Myers Squibb), nimustine (also known as ACNU), and streptozocin (Zanosar, Teva Pharmaceuticals). d. Triazines and Hydrazines
The anti-B7-H3 antibody of the invention may be conjugated to at least one triazine or hydrazine. Triazines and hydrazines are a subclass of nitrogen-containing alkylating agents. In some embodiments, these compounds spontaneously decompose or can be metabolized to produce alkyl diazonium intermediates that facilitate the transfer of an alkyl group to nucleic acids, peptides, and/or polypeptides, thereby causing mutagenic, carcinogenic, or cytotoxic effects. Representative examples include, but are not limited to dacarbazine (DTIC-Dome, Bayer Healthcare Pharmaceuticals Inc.), procarbazine (Mutalane, Sigma-Tau Pharmaceuticals, Inc.), and temozolomide (Temodar, Schering Plough). e. Other Alkylating Agents
The anti-B7-H3 antibodies of the invention may be conjugated to at least one ethylenimine, methylamine derivative, or epoxide. Ethylenimines are a subclass of alkylating agents that typically containing at least one aziridine ring. Epoxides represent a subclass of alkylating agents that are characterized as cyclic ethers with only three ring atoms.
Representatives examples of ethylenimines include, but are not limited to thiopeta (Thioplex, Amgen), diaziquone (also known as aziridinyl benzoquinone (AZQ)), and mitomycin C. Mitomycin C is a natural product that contains an aziridine ring and appears to induce cytoxicity through cross- linking DNA (Dorr RT, et al. Cancer Res.1985;45:3510; Kennedy KA, et al Cancer Res.
1985;45:3541). Representative examples of methylamine derivatives and their analogs include, but are not limited to, altretamine (Hexalen, MGI Pharma, Inc.), which is also known as hexamethylamine and hexastat. Representative examples of epoxides of this class of anti-cancer compound include, but are not limited to dianhydrogalactitol. Dianhydrogalactitol (1,2:5,6-dianhydrodulcitol) is chemically related to the aziridines and generally facilitate the transfer of an alkyl group through a similar mechanism as described above. Dibromodulcitol is hydrolyzed to dianhydrogalactitol and thus is a pro-drug to an epoxide (Sellei C, et al. Cancer Chemother Rep.1969;53:377). 6. Antiangiogenic Agents
In one aspect, the anti-B7-H3 antibodies described herein are conjugated to at least one antiangiogenic agent. Antiangiogenic agents inhibit the growth of new blood vessels. Antiangiogenic agents exert their effects in a variety of ways. In some embodiments, these agents interfere with the ability of a growth factor to reach its target. For example, vascular endothelial growth factor (VEGF) is one of the primary proteins involved in initiating angiogenesis by binding to particular receptors on a cell surface. Thus, certain antiangiogenic agents, that prevent the interaction of VEGF with its cognate receptor, prevent VEGF from initiating angiogenesis. In other embodiments, these agents interfere with intracellular signaling cascades. For example, once a particular receptor on a cell surface has been triggered, a cascade of other chemical signals is initiated to promote the growth of blood vessels. Thus, certain enzymes, for example, some tyrosine kinases, that are known to facilitate intracellular signaling cascades that contribute to, for example, cell proliferation, are targets for cancer treatment. In other embodiments, these agents interfere with intercellular signaling cascades. Yet, in other embodiments, these agents disable specific targets that activate and promote cell growth or by directly interfering with the growth of blood vessel cells. Angiogenesis inhibitory properties have been discovered in more than 300 substances with numerous direct and indirect inhibitory effects.
Representative examples of antiangiogenic agents that may be used in the ADCs of the invention include, but are not limited to, angiostatin, ABX EGF, C1-1033, PKI-166, EGF vaccine, EKB-569, GW2016, ICR-62, EMD 55900, CP358, PD153035, AG1478, IMC-C225 (Erbitux, ZD1839 (Iressa), OSI-774, Erlotinib (tarceva), angiostatin, arrestin, endostatin, BAY 12-9566 and w/fluorouracil or doxorubicin, canstatin, carboxyamidotriozole and with paclitaxel, EMD121974, S- 24, vitaxin, dimethylxanthenone acetic acid, IM862, Interleukin-12, Interleukin-2, NM-3, HuMV833, PTK787, RhuMab, angiozyme (ribozyme), IMC-1C11, Neovastat, marimstat, prinomastat, BMS- 275291,COL-3, MM1270, SU101, SU6668, SU11248, SU5416, with paclitaxel, with gemcitabine and cisplatin, and with irinotecan and cisplatin and with radiation, tecogalan, temozolomide and PEG interferon α2b, tetrathiomolybdate, TNP-470, thalidomide, CC-5013 and with taxotere, tumstatin, 2- methoxyestradiol, VEGF trap, mTOR inhibitors (deforolimus, everolimus (Afinitor, Novartis Pharmaceutical Corporation), and temsirolimus (Torisel, Pfizer, Inc.)), kinase inhibitors (e.g., erlotinib (Tarceva, Genentech, Inc.), imatinib (Gleevec, Novartis Pharmaceutical Corporation), gefitinib (Iressa, AstraZeneca Pharmaceuticals), dasatinib (Sprycel, Brystol-Myers Squibb), sunitinib (Sutent, Pfizer, Inc.), nilotinib (Tasigna, Novartis Pharmaceutical Corporation), lapatinib (Tykerb, GlaxoSmithKline Pharmaceuticals), sorafenib (Nexavar, Bayer and Onyx), phosphoinositide 3- kinases (PI3K), Osimertinib, Cobimetinib, Trametinib, Dabrafenib, Dinaciclib). 7. Antimetabolites
The anti-B7-H3 antibodies of the invention may be conjugated to at least one antimetabolite. Antimetabolites are types of chemotherapy treatments that are very similar to normal substances within the cell. When the cells incorporate an antimetabolite into the cellular metabolism, the result is negative for the cell, e.g., the cell is unable to divide. Antimetabolites are classified according to the substances with which they interfere. Examples of antimetabolites that may be used in the ADCs of the invention include, but are not limited to, a folic acid antagonist (e.g., methotrexate), a pyrimidine antagonist (e.g., 5-Fluorouracil, Foxuridine, Cytarabine, Capecitabine, and Gemcitabine), a purine antagonist (e.g., 6-Mercaptopurine and 6-Thioguanine) and an adenosine deaminase inhibitor (e.g., Cladribine, Fludarabine, Nelarabine and Pentostatin), as described in more detail below. a. Antifolates
The anti-B7-H3 antibodies of the invention may be conjugated to at least one antifolate. Antifolates are a subclass of antimetabolites that are structurally similar to folate. Representative examples include, but are not limited to, methotrexate, 4-amino-folic acid (also known as aminopterin and 4-aminopteroic acid), lometrexol (LMTX), pemetrexed (Alimpta, Eli Lilly and Company), and trimetrexate (Neutrexin, Ben Venue Laboratories, Inc.) b. Purine Antagonists
The anti-B7-H3 antibodies of the invention may be conjugated to at least one purine antagonist. Purine analogs are a subclass of antimetabolites that are structurally similar to the group of compounds known as purines. Representative examples of purine antagonists include, but are not limited to, azathioprine (Azasan, Salix; Imuran, GlaxoSmithKline), cladribine (Leustatin [also known as 2-CdA], Janssen Biotech, Inc.), mercaptopurine (Purinethol [also known as 6-mercaptoethanol], GlaxoSmithKline), fludarabine (Fludara, Genzyme Corporation), pentostatin (Nipent, also known as 2'-deoxycoformycin (DCF)), 6-thioguanine (Lanvis [also known as thioguanine], GlaxoSmithKline). c. Pyrimidine Antagonists
The anti-B7-H3 antibodies of the invention may be conjugated to at least one pyrimidine antagonist. Pyrimidine antagonists are a subclass of antimetabolites that are structurally similar to the group of compounds known as purines. Representative examples of pyrimidine antagonists include, but are not limited to azacitidine (Vidaza, Celgene Corporation), capecitabine (Xeloda, Roche Laboratories), Cytarabine (also known as cytosine arabinoside and arabinosylcytosine, Bedford Laboratories), decitabine (Dacogen, Eisai Pharmaceuticals), 5-fluorouracil (Adrucil, Teva
Pharmaceuticals; Efudex, Valeant Pharmaceuticals, Inc), 5-fluoro-2’-deoxyuridine 5’-phosphate (FdUMP), 5-fluorouridine triphosphate, and gemcitabine (Gemzar, Eli Lilly and Company). 8. Boron-Containing Agents
The anti-B7-H3 antibody of the invention may be conjugated to at least one boron containing agent. Boron-containing agents comprise a class of cancer therapeutic compounds which interfere with cell proliferation. Representative examples of boron containing agents include, but are not limited, to borophycin and bortezomib (Velcade, Millenium Pharmaceuticals). 9. Chemoprotective Agents
The anti-B7-H3 antibodies of the invention may be conjugated to at least one
chemoprotective agent. Chemoprotective drugs are a class of compounds, which help protect the body against specific toxic effects of chemotherapy. Chemoprotective agents may be administered with various chemotherapies in order to protect healthy cells from the toxic effects of chemotherapy drugs, while simultaneously allowing the cancer cells to be treated with the administered
chemotherapeutic Representative chemoprotective agents include but are not limited to amifostine (Ethyol, Medimmune, Inc.), which is used to reduce renal toxicity associated with cumulative doses of cisplatin, dexrazoxane (Totect, Apricus Pharma; Zinecard), for the treatment of extravasation caused by the administration of anthracycline (Totect), and for the treatment of cardiac-related complications caused by the administration of the antitumor antibiotic doxorubicin (Zinecard), and mesna (Mesnex, Bristol-Myers Squibb), which is used to prevent hemorrhagic cystitis during chemotherapy treatment with ifocfamide. 10. Hormone agents
The anti-B7-H3 antibody of the invention may be conjugated to at least one hormone agent. A hormone agent (including synthetic hormones) is a compound that interferes with the production or activity of endogenously produced hormones of the endocrine system. In some embodiments, these compounds interfere with cell growth or produce a cytotoxic effect. Non-limiting examples include androgens, estrogens, medroxyprogesterone acetate (Provera, Pfizer, Inc.), and progestins. 11. Antihormone Agents
The anti-B7-H3 antibodies of the invention may be conjugated to at least one antihormone agent. An“antihormone” agent is an agent that suppresses the production of and/or prevents the function of certain endogenous hormones. In one embodiment, the antihormone agent interferes with the activity of a hormone selected from the group comprising androgens, estrogens, progesterone, and goanadotropin-releasing hormone, thereby interfering with the growth of various cancer cells.
Representative examples of antihormone agents include, but are not limited to, aminoglutethimide, anastrozole (Arimidex, AstraZeneca Pharmaceuticals), bicalutamide (Casodex, AstraZeneca
Pharmaceuticals), cyproterone acetate (Cyprostat, Bayer PLC), degarelix (Firmagon, Ferring
Pharmaceuticals), exemestane (Aromasin, Pfizer Inc.), flutamide (Drogenil, Schering-Plough Ltd), fulvestrant (Faslodex, AstraZeneca Pharmaceuticals), goserelin (Zolodex, AstraZeneca
Pharmaceuticals), letrozole (Femara, Novartis Pharmaceuticals Corporation), leuprolide (Prostap), lupron, medroxyprogesterone acetate (Provera, Pfizer Inc.), Megestrol acetate (Megace, Bristol-Myers Squibb Company), tamoxifen (Nolvadex, AstraZeneca Pharmaceuticals), and triptorelin (Decapetyl, Ferring). 12. Corticosteroids
The anti-B7-H3 antibodies of the invention may be conjugated to at least one corticosteroid. Corticosteroids may be used in the ADCs of the invention to decrease inflammation. An example of a corticosteroid includes, but is not limited to, a glucocorticoid, for example, prednisone (Deltasone, Pharmacia & Upjohn Company, a division of Pfizer, Inc.). 13. Photoactive Therapeutic Agents
The anti-B7-H3 antibodies of the invention may be conjugated to at least one photoactive therapeutic agent. Photoactive therapeutic agents include compounds that can be deployed to kill treated cells upon exposure to electromagnetic radiation of a particular wavelength. Therapeutically relevant compounds absorb electromagnetic radiation at wavelengths which penetrate tissue. In preferred embodiments, the compound is administered in a non-toxic form that is capable of producing a photochemical effect that is toxic to cells or tissue upon sufficient activation. In other preferred embodiments, these compounds are retained by cancerous tissue and are readily cleared from normal tissues. Non-limiting examples include various chromagens and dyes. 14. Oligonucleotides
The anti-B7-H3 antibodies of the invention may be conjugated to at least one oligonucleotide. Oligonucleotides are made of short nucleic acid chains that work by interfering with the processing of genetic information. In some embodiments, the oligonucleotides for use in ADCs are unmodified single-stranded and/or double-stranded DNA or RNA molecules, while in other embodiments, these therapeutic oligonucleotides are chemically-modified single-stranded and/or double-stranded DNA or RNA molecules. In one embodiment, the oligonulceotides used in the ADCs are relatively short (19– 25 nucleotides) and hybridize to a unique nucleic acid sequence in the total pool of nucleic acid targets present in cells. Some of the important oligonucleotide technologies include the antisense oligonucleotides (including RNA interference (RNAi)), aptamers, CpG oligonucleotides, and ribozymes. a. Antisense oligonucleotides
The anti-B7-H3 antibody of the invention may be conjugated to at least one antisense oligonucleotide. Antisense oligonucleotides are designed to bind to RNA through Watson–Crick hybridization. In some embodiments the antisense oligonucleotide is complementary to a nucleotide encoding a region, domain, portion, or segment of B7-H3. In some embodiments, the antisense oligonucleotide comprises from about 5 to about 100 nucleotides, from about 10 to about 50 nucleotides, from about 12 to about 35, and from about 18 to about 25 nucleotides. In some embodiments, the oligonucleotide is at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 100% homologous to a region, portion, domain, or segment of the B7-H3 gene. In some embodiments there is substantial sequence homology over at least 15, 20, 25, 30, 35, 40, 50, or 100 consecutive nucleotides of the B7- H3 gene. In preferred embodiments, the size of these antisense oligonucleotides ranges from 12 to 25 nucleotides in length, with the majority of antisense oligonucleotides being 18 to 21 nucleotides in length. There are multiple mechanisms that can be exploited to inhibit the function of the RNA once the oligonucleotide binds to the target RNA (Crooke ST. (1999). Biochim. Biophys. Acta, 1489, 30– 42). The best-characterized antisense mechanism results in cleavage of the targeted RNA by endogenous cellular nucleases, such as RNase H or the nuclease associated with the RNA interference mechanism. However, oligonucleotides that inhibit expression of the target gene by non-catalytic mechanisms, such as modulation of splicing or translation arrest, can also be potent and selective modulators of gene function.
Another RNase-dependent antisense mechanism that has recently received much attention is RNAi (Fire et al. (1998). Nature, 391, 806–811.; Zamore PD. (2002). Science, 296, 1265–1269.). RNA interference (RNAi) is a post-transcriptional process where a double stranded RNA inhibits gene expression in a sequence specific fashion. In some embodiments, the RNAi effect is achieved through the introduction of relatively longer double-stranded RNA (dsRNA), while in preferred embodiments, this RNAi effect is achieved by the introduction of shorter double-stranded RNAs, e.g. small interfering RNA (siRNA) and/or microRNA (miRNA). In yet another embodiment, RNAi can also be achieved by introducing of plasmid that generate dsRNA complementary to target gene. In each of the foregoing embodiments, the double-stranded RNA is designed to interfere with the gene expression of a particular the target sequence within cells. Generally, the mechanism involves conversion of dsRNA into short RNAs that direct ribonucleases to homologous mRNA targets (summarized, Ruvkun, Science 2294:797 (2001)), which then degrades the corresponding endogenous mRNA, thereby resulting in the modulation of gene expression. Notably, dsRNA has been reported to have anti-proliferative properties, which makes it possible also to envisage therapeutic applications (Aubel et al., Proc. Natl. Acad. Sci., USA 88:906 (1991)). For example, synthetic dsRNA has been shown to inhibit tumor growth in mice (Levy et al. Proc. Nat. Acad. Sci. USA, 62:357-361 (1969)), is active in the treatment of leukemic mice (Zeleznick et al., Proc. Soc. Exp. Biol. Med.130:126-128 (1969)), and inhibits chemically induced tumorigenesis in mouse skin (Gelboin et al., Science 167:205-207 (1970)). Thus, in a preferred embodiment, the invention provides for the use of antisense oligonucleotides in ADCs for the treatment of breast cancer. In other embodiments, the invention provides compositions and methods for initiating antisense oligonucleotide treatment, wherein dsRNA interferes with target cell expression of B7-H3 at the mRNA level. dsRNA, as used above, refers to naturally-occurring RNA, partially purified RNA, recombinantly produced RNA, synthetic RNA, as well as altered RNA that differs from naturally-occurring RNA by the inclusion of non-standard nucleotides, non-nucleotide material, nucleotide analogs (e.g. locked nucleic acid (LNA)), deoxyribonucleotides, and any combination thereof. RNA of the invention need only be sufficiently similar to natural RNA that it has the ability to mediate the antisense oligonucleotide- based modulation described herein. b. Aptamers
The anti-B7-H3 antibodies of the invention may be conjugated to at least one aptamer. An aptamer is a nucleic acid molecule that has been selected from random pools based on its ability to bind other molecules. Like antibodies, aptamers can bind target molecules with extraordinary affinity and specificity In many embodiments aptamers assume complex sequence-dependent three- dimensional shapes that allow them to interact with a target protein, resulting in a tightly bound complex analogous to an antibody-antigen interaction, thereby interfering with the function of said protein. The particular capacity of aptamers to bind tightly and specifically to their target protein underlines their potential as targeted molecular therapies. c. CpG oligonucleotides
The anti-B7-H3 antibodies of the invention may be conjugated to at least one CpG oligonucleotide. Bacterial and viral DNA are known to be a strong activators of both the innate and specific immunity in humans. These immunologic characteristics have been associated with unmethylated CpG dinucleotide motifs found in bacterial DNA. Owing to the fact that these motifs are rare in humans, the human immune system has evolved the ability to recognize these motifs as an early indication of infection and subsequently initiate immune responses. Therefore, oligonucleotides containing this CpG motif can be exploited to initiate an antitumor immune response. d. Ribozymes
The anti-B7-H3 antibody of the invention may be conjugated to at least one ribozyme.
Ribozymes are catalytic RNA molecules ranging from about 40 to 155 nucleotides in length. The ability of ribozymes to recognize and cut specific RNA molecules makes them potential candidates for therapeutics. A representative example includes angiozyme. 15. Radionuclide Agents (Radioactive Isotopes)
The anti-B7-H3 antibodies of the invention may be conjugated to at least one radionuclide agent. Radionuclide agents comprise agents that are characterized by an unstable nucleus that is capable of undergoing radioactive decay. The basis for successful radionuclide treatment depends on sufficient concentration and prolonged retention of the radionuclide by the cancer cell. Other factors to consider include the radionuclide half-life, the energy of the emitted particles, and the maximum range that the emitted particle can travel. In preferred embodiments, the therapeutic agent is a radionuclide selected from the group consisting of 111In, 177Lu, 212Bi, 213Bi, 211At, 62Cu, 64Cu, 67Cu, 90Y, I25I, I31I, 32P, 33P, 47Sc, 111Ag, 67Ga, 142Pr, 153Sm, 161Tb, 166Dy, 166Ho, 186Re, 188Re, 189Re, 212Pb, 223Ra, 225Ac, 59Fe, 75Se, 77As, 89Sr, 99Mo, 105Rh, I09Pd, 143Pr, 149Pm, 169Er, 194Ir, 198Au, 199Au, and 211Pb. Also preferred are radionuclides that substantially decay with Auger-emitting particles. For example, Co- 58, Ga-67, Br-80m, Tc-99m, Rh-103m, Pt-109, In-1111, Sb-119, I-125, Ho-161, Os-189m and Ir- 192. Decay energies of useful beta-particle-emitting nuclides are preferably Dy-152, At-211, Bi-212, Ra-223, Rn-219, Po-215, Bi-211, Ac-225, Fr-221, At-217, Bi-213 and Fm-255. Decay energies of useful alpha-particle-emitting radionuclides are preferably 2,000-10,000 keV, more preferably 3,000- 8,000 keV, and most preferably 4,000-7,000 keV. Additional potential radioisotopes of use include 11C 13N 150 75Br 198Au 224Ac 126I 133I 77Br 113mIn, 95Ru, 97Ru, I03Ru, 105Ru, 107Hg, 203Hg, 121mTe,122mTe, 125mTe, 165Tm, I67Tm, 168Tm, 197Pt, 109Pd, 105Rh, 142Pr, 143Pr, 161Tb, !66Ho, 199Au, 57Co, 58Co, 51Cr, 59Fe, 75Se, 201Tl, 225Ac, 76Br, I69Yb, and the like. 16. Radiosensitizers
The anti-B7-H3 antibodies of the invention may be conjugated to at least one radiosensitizer. The term“radiosensitizer,” as used herein, is defined as a molecule, preferably a low molecular weight molecule, administered to animals in therapeutically effective amounts to increase the sensitivity of the cells to be radiosensitized to electromagnetic radiation and/or to promote the treatment of diseases that are treatable with electromagnetic radiation. Radiosensitizers are agents that make cancer cells more sensitive to radiation therapy, while typically having much less of an effect on normal cells. Thus, the radiosensitizer can be used in combination with a radiolabeled antibody or ADC. The addition of the radiosensitizer can result in enhanced efficacy when compared to treatment with the radiolabeled antibody or antibody fragment alone. Radiosensitizers are described in D. M. Goldberg (ed.), Cancer Therapy with Radiolabeled Antibodies, CRC Press (1995). Examples of radiosensitizers include gemcitabine, 5-fluorouracil, taxane, and cisplatin.
Radiosensitizers may be activated by the electromagnetic radiation of X-rays. Representative examples of X-ray activated radiosensitizers include, but are not limited to, the following:
metronidazole, misonidazole, desmethylmisonidazole, pimonidazole, etanidazole, nimorazole, mitomycin C, RSU 1069, SR 4233, E09, RB 6145, nicotinamide, 5-bromodeoxyuridine (BUdR), 5- iododeoxyuridine (IUdR), bromodeoxycytidine, fluorodeoxyuridine (FUdR), hydroxyurea, cisplatin, and therapeutically effective analogs and derivatives of the same. Alternatively, radiosensitizers may be activated using photodynamic therapy (PDT). Representative examples of photodynamic radiosensitizers include, but are not limited to, hematoporphyrin derivatives, Photofrin(r), benzoporphyrin derivatives, NPe6, tin etioporphyrin (SnET2), pheoborbide a, bacteriochlorophyll a, naphthalocyanines, phthalocyanines, zinc phthalocyanine, and therapeutically effective analogs and derivatives of the same. 16. Topoisomerase Inhibitors
The anti-B7-H3 antibodies of the invention may be conjugated to at least one topoisomerase inhibitor. Topoisomerase inhibitors are chemotherapy agents designed to interfere with the action of topoisomerase enzymes (topoisomerase I and II), which are enzymes that control the changes in DNA structure by catalyzing then breaking and rejoining of the phosphodiester backbone of DNA strands during the normal cell cycle. Representative examples of DNA topoisomerase I inhibitors include, but are not limited to, camptothecins and its derivatives irinotecan (CPT-11, Camptosar, Pfizer, Inc.) and topotecan (Hycamtin, GlaxoSmithKline Pharmaceuticals). Representative examples of DNA topoisomerase II inhibitors include, but are not limited to, amsacrine, daunorubicin, doxotrubicin, epipodophyllotoxins, ellipticines, epirubicin, etoposide, razoxane, and teniposide. 17. Kinase Inhibitors
The anti-B7-H3 antibodies of the invention may be conjugated to at least one kinase inhibitor. By blocking the ability of protein kinases to function, tumor growth may be inhibited. Examples of kinase inhibitors that may be used in the ADCs of the invention include, but are not limited to, Axitinib, Bosutinib, Cediranib, Dasatinib, Erlotinib, Gefitinib, Imatinib, Lapatinib, Lestaurtinib, Nilotinib, Semaxanib, Sunitinib, Osimertinib, Cobimetinib, Trametinib, Dabrafenib, Dinaciclib, and Vandetanib. 18. Other Agents
Examples of other agents that may be used in the ADCs of the invention include, but are not limited to, abrin (e.g. abrin A chain), alpha toxin, Aleurites fordii proteins, amatoxin, crotin, curcin, dianthin proteins, diptheria toxin (e.g. diphtheria A chain and nonbinding active fragments of diphtheria toxin), deoxyribonuclease (Dnase), gelonin, mitogellin, modeccin A chain, momordica charantia inhibitor, neomycin, onconase, phenomycin, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), pokeweed antiviral protein, Pseudomonas endotoxin, Pseudomonas exotoxin (e.g.
exotoxin A chain (from Pseudomonas aeruginosa)), restrictocin, ricin A chain, ribonuclease (Rnase), sapaonaria officinalis inhibitor, saporin, alpha-sarcin, Staphylcoccal enterotoxin-A, tetanus toxin, cisplatin, carboplatin, and oxaliplatin (Eloxatin, Sanofi Aventis), proteasome inhibitors (e.g. PS-341 [bortezomib or Velcade]), HDAC inhibitors (vorinostat (Zolinza, Merck & Company, Inc.)), belinostat, entinostat, mocetinostat, and panobinostat), COX-2 inhibitors, substituted ureas, heat shock protein inhibitors (e.g. Geldanamycin and its numerous analogs), adrenocortical suppressants, and the tricothecenes. (See, for example, WO 93/21232). Other agents also include asparaginase (Espar, Lundbeck Inc.), hydroxyurea, levamisole, mitotane (Lysodren, Bristol-Myers Squibb), and tretinoin (Renova, Valeant Pharmaceuticals Inc.). III.C. Anti-B7-H3 ADCs: Other Exemplary Linkers
In addition to the linkers mentioned above, other exemplary linkers include, but are not limited to, 6-maleimidocaproyl, maleimidopropanoyl (“MP”), valine-citrulline (“val-cit” or“vc”), alanine- phenylalanine (“ala-phe”), p-aminobenzyloxycarbonyl (a“PAB”), N-Succinimidyl 4-(2-pyridylthio) pentanoate (“SPP”), and 4-(N-maleimidomethyl)cyclohexane-1 carboxylate (“MCC”).
In one aspect, an anti-B7-H3 antibody is conjugated to a drug, (such as auristatin, e.g., MMAE), via a linker comprising maleimidocaproyl (“mc”), valine citrulline (val-cit or“vc”), and PABA (referred to as a“mc-vc-PABA linker”). Maleimidocaproyl acts as a linker to the anti-B7-H3 antibody and is not cleavable. Val-cit is a dipeptide that is an amino acid unit of the linker and allows for cleavage of the linker by a protease, specifically the protease cathepsin B. Thus, the val-cit component of the linker provides a means for releasing the auristatin from the ADC upon exposure to the intracellular environment Within the linker p-aminobenzylalcohol (PABA) acts as a spacer and is self immolative, allowing for the release of the MMAE. The structure of the mc-vc-PABA-MMAE linker is provided in Figure 3.
As described above, suitable linkers include, for example, cleavable and non-cleavable linkers. A linker may be a“cleavable linker,” facilitating release of a drug. Nonlimiting exemplary cleavable linkers include acid-labile linkers (e.g., comprising hydrazone), protease-sensitive (e.g., peptidase-sensitive) linkers, photolabile linkers, or disulfide-containing linkers (Chari et al., Cancer Research 52:127-131 (1992); U.S. Pat. No.5,208,020). A cleavable linker is typically susceptible to cleavage under intracellular conditions. Suitable cleavable linkers include, for example, a peptide linker cleavable by an intracellular protease, such as lysosomal protease or an endosomal protease. In exemplary embodiments, the linker can be a dipeptide linker, such as a valine-citrulline (val-cit) or a phenylalanine-lysine (phe-lys) linker.
Linkers are preferably stable extracellularly in a sufficient manner to be therapeutically effective. Before transport or delivery into a cell, the ADC is preferably stable and remains intact, i.e. the antibody remains conjugated to the drug moiety. Linkers that are stable outside the target cell may be cleaved at some efficacious rate once inside the cell. Thus, an effective linker will: (i) maintain the specific binding properties of the antibody; (ii) allow delivery, e.g., intracellular delivery, of the drug moiety; and (iii) maintain the therapeutic effect, e.g., cytotoxic effect, of a drug moiety.
In one embodiment, the linker is cleavable under intracellular conditions, such that cleavage of the linker sufficiently releases the drug from the antibody in the intracellular environment to be therapeutically effective. In some embodiments, the cleavable linker is pH-sensitive, i.e., sensitive to hydrolysis at certain pH values. Typically, the pH-sensitive linker is hydrolyzable under acidic conditions. For example, an acid-labile linker that is hydrolyzable in the lysosome (e.g., a hydrazone, semicarbazone, thiosemicarbazone, cis-aconitic amide, orthoester, acetal, ketal, or the like) can be used. (See, e.g., U.S. Pat. Nos.5,122,368; 5,824,805; 5,622,929; Dubowchik and Walker, 1999, Pharm. Therapeutics 83:67-123; Neville et al., 1989, Biol. Chem.264:14653-14661.) Such linkers are relatively stable under neutral pH conditions, such as those in the blood, but are unstable at below pH 5.5 or 5.0, the approximate pH of the lysosome. In certain embodiments, the hydrolyzable linker is a thioether linker (such as, e.g., a thioether attached to the therapeutic agent via an acylhydrazone bond (see, e.g., U.S. Pat. No.5,622,929).
In other embodiments, the linker is cleavable under reducing conditions (e.g., a disulfide linker). A variety of disulfide linkers are known in the art, including, for example, those that can be formed using SATA (N-succinimidyl-5-acetylthioacetate), SPDP (N-succinimidyl-3-(2- pyridyldithio)propionate), SPDB (N-succinimidyl-3-(2-pyridyldithio)butyrate) and SMPT (N- succinimidyloxycarbonyl-alpha-methyl-alpha-(2-pyridyl-dithio)toluene), SPDB and SMPT. (See, e.g., Thorpe et al., 1987, Cancer Res.47:5924-5931; Wawrzynczak et al., In Immunoconjugates: Antibody Conjugates in Radioimagery and Therapy of Cancer (C. W. Vogel ed., Oxford U. Press, 1987. See also U S Pat No 4880935 ) In some embodiments, the linker is cleavable by a cleaving agent, e.g., an enzyme, that is present in the intracellular environment (e.g., within a lysosome or endosome or caveolea). The linker can be, e.g., a peptidyl linker that is cleaved by an intracellular peptidase or protease enzyme, including, but not limited to, a lysosomal or endosomal protease. In some embodiments, the peptidyl linker is at least two amino acids long or at least three amino acids long. Cleaving agents can include cathepsins B and D and plasmin, all of which are known to hydrolyze dipeptide drug derivatives resulting in the release of active drug inside target cells (see, e.g., Dubowchik and Walker, 1999, Pharm. Therapeutics 83:67-123). Most typical are peptidyl linkers that are cleavable by enzymes that are present in B7-H3-expressing cells. Examples of such linkers are described, e.g., in U.S. Pat. No. 6,214,345, incorporated herein by reference in its entirety and for all purposes. In a specific embodiment, the peptidyl linker cleavable by an intracellular protease is a Val-Cit linker or a Phe-Lys linker (see, e.g., U.S. Pat. No.6,214,345, which describes the synthesis of doxorubicin with the val-cit linker). One advantage of using intracellular proteolytic release of the therapeutic agent is that the agent is typically attenuated when conjugated and the serum stabilities of the conjugates are typically high.
In other embodiments, the linker is a malonate linker (Johnson et al., 1995, Anticancer Res. 15:1387-93), a maleimidobenzoyl linker (Lau et al., 1995, Bioorg-Med-Chem.3(10):1299-1304), or a 3'-N-amide analog (Lau et al., 1995, Bioorg-Med-Chem.3(10): 1305-12).
In yet other embodiments, the linker unit is not cleavable and the drug is released, for example, by antibody degradation. See U.S. Publication No.20050238649 incorporated by reference herein in its entirety. An ADC comprising a non-cleavable linker may be designed such that the ADC remains substantially outside the cell and interacts with certain receptors on a target cell surface such that the binding of the ADC initiates (or prevents) a particular cellular signaling pathway.
In some embodiments, the linker is substantially hydrophilic linker (e.g., PEG4Mal and sulfo- SPDB). A hydrophilic linker may be used to reduce the extent to which the drug may be pumped out of resistant cancer cells through MDR (multiple drug resistance) or functionally similar transporters.
In other embodiments, upon cleavage, the linker functions to directly or indirectly inhibit cell growth and/or cell proliferation. For example, in some embodiments, the linker, upon cleavage, can function as an intercalating agent, thereby inhibiting macromolecular biosynthesis (e.g. DNA replication, RNA transcription, and/or protein synthesis).
In other embodiments, the linker is designed to facilitate bystander killing (the killing of neighboring cells) through diffusion of the linker-drug and/or the drug alone to neighboring cells. In other, embodiments, the linker promotes cellular internalization.
The presence of a sterically hindered disulfide can increase the stability of a particular disulfide bond, enhancing the potency of the ADC. Thus, in one embodiment, the linker includes a sterically hindered disulfide linkage. A sterically hindered disulfide refers to a disulfide bond present within a particular molecular environment wherein the environment is characterized by a particular spatial arrangement or orientation of atoms, typically within the same molecule or compound, which prevents or at least partially inhibits the reduction of the disulfide bond. Thus, the presence of bulky (or sterically hindering) chemical moieties and/or bulky amino acid side chains proximal to the disulfide bond prevents or at least partially inhibits the disulfide bond from potential interactions that would result in the reduction of the disulfide bond.
Notably, the aforementioned linker types are not mutually exclusive. For example, in one embodiment, the linker used in the anti-B7-H3 ADCs described herein is a non-cleavable linker that promotes cellular internalization.
In some embodiments, a linker component comprises a“stretcher unit” that links an antibody to another linker component or to a drug moiety. An illustrative stretcher unit described in U.S.
8,309,093, incorporated by reference herein. In certain embodiments, the stretcher unit is linked to the anti-B7-H3 antibody via a disulfide bond between a sulfur atom of the anti-B7-H3 antibody unit and a sulfur atom of the stretcher unit. A representative stretcher unit of this embodiment is depicted in U.S.8,309,093, incorporated by reference herein. In yet other embodiments, the stretcher contains a reactive site that can form a bond with a primary or secondary amino group of an antibody.
Examples of these reactive sites include but are not limited to, activated esters such as succinimide esters, 4 nitrophenyl esters, pentafluorophenyl esters, tetrafluorophenyl esters, anhydrides, acid chlorides, sulfonyl chlorides, isocyanates and isothiocyanates. Representative stretcher units of this embodiment are depicted in U.S.8,309,093, incorporated by reference herein.
In some embodiments, the stretcher contains a reactive site that is reactive to a modified carbohydrate's (—CHO) group that can be present on an antibody. For example, a carbohydrate can be mildly oxidized using a reagent such as sodium periodate and the resulting (—CHO) unit of the oxidized carbohydrate can be condensed with a Stretcher that contains a functionality such as a hydrazide, an oxime, a primary or secondary amine, a hydrazine, a thiosemicarbazone, a hydrazine carboxylate, and an arylhydrazide such as those described by Kaneko et al., 1991, Bioconjugate Chem.2:133-41. Representative Stretcher units of this embodiment are depicted in U.S.8,309,093, incorporated by reference herein.
In some embodiments, a linker component comprises an“amino acid unit”. In some such embodiments, the amino acid unit allows for cleavage of the linker by a protease, thereby facilitating release of the drug from the immunoconjugate upon exposure to intracellular proteases, such as lysosomal enzymes (Doronina et al. (2003) Nat. Biotechnol.21:778-784). Exemplary amino acid units include, but are not limited to, dipeptides, tripeptides, tetrapeptides, and pentapeptides.
Exemplary dipeptides include, but are not limited to, valine-citrulline (vc or val-cit), alanine- phenylalanine (af or ala-phe); phenylalanine-lysine (fk or phe-lys); phenylalanine-homolysine (phe- homolys); and N-methyl-valine-citrulline (Me-val-cit). Exemplary tripeptides include, but are not limited to, glycine-valine-citrulline (gly-val-cit) and glycine-glycine-glycine (gly-gly-gly). An amino acid unit may comprise amino acid residues that occur naturally and/or minor amino acids and/or non- naturally occurring amino acid analogs, such as citrulline Amino acid units can be designed and optimized for enzymatic cleavage by a particular enzyme, for example, a tumor-associated protease, cathepsin B, C and D, or a plasmin protease.
In one embodiment, the amino acid unit is valine-citrulline (vc or val-cit). In another aspect, the amino acid unit is phenylalanine-lysine (i.e., fk). In yet another aspect of the amino acid unit, the amino acid unit is N-methylvaline-citrulline. In yet another aspect, the amino acid unit is 5- aminovaleric acid, homo phenylalanine lysine, tetraisoquinolinecarboxylate lysine, cyclohexylalanine lysine, isonepecotic acid lysine, beta-alanine lysine, glycine serine valine glutamine and isonepecotic acid.
Alternatively, in some embodiments, the amino acid unit is replaced by a glucuronide unit that links a stretcher unit to a spacer unit if the stretcher and spacer units are present, links a stretcher unit to the drug moiety if the spacer unit is absent, and links the linker unit to the drug if the stretcher and spacer units are absent. The glucuronide unit includes a site that can be cleaved by a β- glucuronidase enzyme (See also US 2012/0107332, incorporated by reference herein). In some embodiments, the glucuronide unit comprises a sugar moiety (Su) linked via a glycoside bond (— O'—) to a self-immolative group (Z) of the formula as depicted below (See also US 2012/0107332, incorporated by reference herein).
Figure imgf000281_0001
The glycosidic bond (—O'—) is typically a β-glucuronidase-cleavage site, such as a bond cleavable by human, lysosomal β-glucuronidase. In the context of a glucuronide unit, the term“self-immolative group” refers to a di- or tri-functional chemical moiety that is capable of covalently linking together two or three spaced chemical moieties (i.e., the sugar moiety (via a glycosidic bond), a drug moiety (directly or indirectly via a spacer unit), and, in some embodiments, a linker (directly or indirectly via a stretcher unit) into a stable molecule. The self-immolative group will spontaneously separate from the first chemical moiety (e.g., the spacer or drug unit) if its bond to the sugar moiety is cleaved.
In some embodiments, the sugar moiety (Su) is cyclic hexose, such as a pyranose, or a cyclic pentose, such as a furanose. In some embodiments, the pyranose is a glucuronide or hexose. The sugar moiety is usually in the β-D conformation. In a specific embodiment, the pyranose is a β-D- glucuronide moiety (i.e., β-D-glucuronic acid linked to the self-immolative group—Z— via a glycosidic bond that is cleavable by β-glucuronidase). In some embodiments, the sugar moiety is unsubstituted (e.g., a naturally occurring cyclic hexose or cyclic pentose). In other embodiments, the sugar moiety can be a substituted β-D-glucuronide (i.e., glucuronic acid substituted with one or more group, such hydrogen, hydroxyl, halogen, sulfur, nitrogen or lower alkyl. In some embodiments, the glucuronide unit has one of the formulas as described in US 2012/0107332, incorporated by reference herein.
In some embodiments, the linker comprises a spacer unit (—Y—), which, when present, links an amino acid unit (or Glucuronide unit, see also US 2012/0107332, incorporated by reference herein) to the drug moiety when an amino acid unit is present. Alternately, the spacer unit links the stretcher unit to the drug moiety when the amino acid unit is absent. The spacer unit may also links the drug unit to the antibody unit when both the amino acid unit and stretcher unit are absent.
Spacer units are of two general types: non self-immolative or self-immolative. A non self- immolative spacer unit is one in which part or all of the spacer unit remains bound to the drug moiety after cleavage, particularly enzymatic, of an amino acid unit (or glucuronide unit) from the antibody- drug conjugate. Examples of a non self-immolative spacer unit include, but are not limited to a (glycine-glycine) spacer unit and a glycine spacer unit (see U.S.8,309,093, incorporated by reference herein)).Other examples of self-immolative spacers include, but are not limited to, aromatic compounds that are electronically similar to the PAB group such as 2-aminoimidazol-5-methanol derivatives (Hay et al., 1999, Bioorg. Med. Chem. Lett.9:2237) and ortho or para- aminobenzylacetals. Spacers can be used that undergo cyclization upon amide bond hydrolysis, such as substituted and unsubstituted 4-aminobutyric acid amides (Rodrigues et al., 1995, Chemistry Biology 2:223), appropriately substituted bicyclo[2.2.1] and bicyclo[2.2.2] ring systems (Storm et al., 1972, J. Amer. Chem. Soc.94:5815) and 2-aminophenylpropionic acid amides (Amsberry et al., 1990, J. Org. Chem.55:5867). Elimination of amine-containing drugs that are substituted at the α- position of glycine (Kingsbury et al., 1984, J. Med. Chem.27:1447) are also examples of self- immolative spacers..
Other examples of self-immolative spacers include, but are not limited to, aromatic compounds that are electronically similar to the PAB group such as 2-aminoimidazol-5-methanol derivatives (see, e.g., Hay et al., 1999, Bioorg. Med. Chem. Lett.9:2237) and ortho or para- aminobenzylacetals. Spacers can be used that undergo cyclization upon amide bond hydrolysis, such as substituted and unsubstituted 4-aminobutyric acid amides (see, e.g., Rodrigues et al., 1995, Chemistry Biology 2:223), appropriately substituted bicyclo[2.2.1] and bicyclo[2.2.2] ring systems (see, e.g., Storm et al., 1972, J. Amer. Chem. Soc.94:5815) and 2-aminophenylpropionic acid amides (see, e.g., Amsberry et al., 1990, J. Org. Chem.55:5867). Elimination of amine-containing drugs that are substituted at the a-position of glycine (see, e.g., Kingsbury et al., 1984, J. Med. Chem.27:1447) are also examples of self-immolative spacers.
Other suitable spacer units are disclosed in Published U.S. Patent Application No.2005- 0238649, the disclosure of which is incorporated by reference herein.
Another approach for the generation of ADCs involves the use of heterobifunctional cross- linkers which link the anti-B7-H3 antibody to the drug moiety. Examples of cross-linkers that may be used include N-succinimidyl 4-(5-nitro-2-pyridyldithio)-pentanoate or the highly water-soluble analog N-sulfosuccinimidyl 4-(5-nitro-2-pyridyldithio)-pentanoate, N-succinimidyl-4-(2-pyridyldithio) butyrate (SPDB), N-succinimidyl-4-(5-nitro-2-pyridyldithio) butyrate (SNPB), and N- sulfosuccinimidyl-4-(5-nitro-2-pyridyldithio) butyrate (SSNPB), N-succinimidyl-4-methyl-4-(5-nitro- 2-pyridyldithio)pentanoate (SMNP), N-succinimidyl-4-(5-N,N-dimethylcarboxamido-2-pyridyldithio) butyrate (SCPB) or N-sulfosuccinimidyl4-(5-N,N-dimethylcarboxamido-2-pyridyldithio) butyrate (SSCPB)). The antibodies of the invention may be modified with the cross-linkers N-succinimidyl 4- (5-nitro-2-pyridyldithio)-pentanoate, N-sulfosuccinimidyl 4-(5-nitro-2-pyridyldithio)-pentanoate, SPDB, SNPB, SSNPB, SMNP, SCPB, or SSCPB can then react with a small excess of a particular drug that contains a thiol moiety to give excellent yields of an ADC. Preferably, the cross-linkers are compounds of the formula as depicted in U.S. Patent No.6,913,748, incorporated by reference herein.
In one embodiment, charged linkers (also referred to as pro-charged linkers) are used to conjugate anti-B7-H3 antibodies to drugs to form ADCs. Charged linkers include linkers that become charged after cell processing. The presence of a charged group(s) in the linker of a particular ADC or on the drug after cellular processing provides several advantages, such as (i) greater water solubility of the ADC, (ii) ability to operate at a higher concentration in aqueous solutions, (iii) ability to link a greater number of drug molecules per antibody, potentially resulting in higher potency, (iv) potential for the charged conjugate species to be retained inside the target cell, resulting in higher potency, and (v) improved sensitivity of multidrug resistant cells, which would be unable to export the charged drug species from the cell. Examples of some suitable charged or pro-charged cross-linkers and their synthesis are shown in Figures 1 to 10 of U.S. Patent No.8,236, 319, and are incorporated by reference herein. Preferably, the charged or pro-charged cross-linkers are those containing sulfonate, phosphate, carboxyl or quaternary amine substituents that significantly increase the solubility of the ADCs, especially for ADCs with 2 to 20 conjugated drugs. Conjugates prepared from linkers containing a pro-charged moiety would produce one or more charged moieties after the conjugate is metabolized in a cell.
Additional examples of linkers that can be used with the compositions and methods include valine-citrulline; maleimidocaproyl; amino benzoic acids; p-aminobenzylcarbamoyl (PAB);
lysosomal enzyme-cleavable linkers; maleimidocaproyl-polyethylene glycol (MC(PEG)6-OH); N- methyl-valine citrulline; N-succinimidyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC); N-Succinimidyl 4-(2-pyridyldithio)butanoate (SPDB); and N-Succinimidyl 4-(2- pyridylthio)pentanoate (SPP) (See also US 2011/0076232). Another linker for use in the invention includes an avidin-biotin linkage to provide an avidin-biotin-containing ADC (See also U.S. Patent No.4,676,980, PCT publication Nos. WO1992/022332A2, WO1994/016729A1, WO1995/015770A1, WO1997/031655A2, WO1998/035704A1, WO1999/019500A1, WO2001/09785A2,
WO2001/090198A1, WO2003/093793A2, WO2004/050016A2, WO2005/081898A2,
WO2006/083562A2, WO2006/089668A1, WO2007/150020A1, WO2008/135237A1,
WO2010/111198A1 WO2011/057216A1 WO2011/058321A1 WO2012/027494A1 and EP77671B1), wherein some such linkers are resistant to biotinidase cleavage. Additional linkers that may be used in the invention include a cohesin/dockerin pair to provide a cohesion-dockerin- containing ADC (See PCT publication Nos. WO2008/097866A2, WO2008/097870A2,
WO2008/103947A2, and WO2008/103953A2).
Additional linkers for use in the invention may contain non-peptide polymers (examples include, but are not limited to, polyethylene glycol, polypropylene glycol, polyoxyethylated polyols, polyvinyl alcohol, polysaccharides, dextran, polyvinyl ethyl ether, PLA (poly(lactic acid)), PLGA (poly(lactic acid-glycolic acid)), and combinations thereof, wherein a preferred polymer is polyethylene glycol) (See also PCT publication No. WO2011/000370). Additional linkers are also described in WO 2004-010957, U.S. Publication No.20060074008, U.S. Publication No.
20050238649, and U.S. Publication No.20060024317, each of which is incorporated by reference herein in its entirety).
For an ADC comprising a maytansinoid, many positions on maytansinoids can serve as the position to chemically link the linking moiety. In one embodiment, maytansinoids comprise a linking moiety that contains a reactive chemical group are C-3 esters of maytansinol and its analogs where the linking moiety contains a disulfide bond and the chemical reactive group comprises a N-succinimidyl or N-sulfosuccinimidyl ester. For example, the C-3 position having a hydroxyl group, the C-14 position modified with hydroxymethyl, the C-15 position modified with hydroxy and the C-20 position having a hydroxy group are all useful. The linking moiety most preferably is linked to the C- 3 position of maytansinol.
The conjugation of the drug to the antibody via a linker can be accomplished by any technique known in the art. A number of different reactions are available for covalent attachment of drugs and linkers to antibodies. This may be accomplished by reaction of the amino acid residues of the antibody, including the amine groups of lysine, the free carboxylic acid groups of glutamic and aspartic acid, the sulfhydryl groups of cysteine and the various moieties of the aromatic amino acids. One of the most commonly used non-specific methods of covalent attachment is the carbodiimide reaction to link a carboxy (or amino) group of a compound to amino (or carboxy) groups of the antibody. Additionally, bifunctional agents such as dialdehydes or imidoesters have been used to link the amino group of a compound to amino groups of an antibody. Also available for attachment of drugs to antibodies is the Schiff base reaction. This method involves the periodate oxidation of a drug that contains glycol or hydroxy groups, thus forming an aldehyde which is then reacted with the binding agent. Attachment occurs via formation of a Schiff base with amino groups of the antibody. Isothiocyanates can also be used as coupling agents for covalently attaching drugs to antibodies. Other techniques are known to the skilled artisan and within the scope of the invention.
In certain embodiments, an intermediate, which is the precursor of the linker, is reacted with the drug under appropriate conditions. In certain embodiments, reactive groups are used on the drug or the intermediate The product of the reaction between the drug and the intermediate or the derivatized drug, is subsequently reacted with the anti-B7-H3 antibody under appropriate conditions. The synthesis and structure of exemplary linkers, stretcher units, amino acid units, self-immolative spacer units are described in U.S. Patent Application Publication Nos.20030083263, 20050238649 and 20050009751, each if which is incorporated herein by reference.
Stability of the ADC may be measured by standard analytical techniques such as mass spectroscopy, HPLC, and the separation/analysis technique LC/MS. IV. Purification of Anti-B7-H3 ADCs
Purification of the ADCs may be achieved in such a way that ADCs having certain DARs are collected. For example, HIC resin may be used to separate high drug loaded ADCs from ADCs having optimal drug to antibody ratios (DARs), e.g. a DAR of 4 or less. In one embodiment, a hydrophobic resin is added to an ADC mixture such that undesired ADCs, i.e., higher drug loaded ADCs, bind the resin and can be selectively removed from the mixture. In certain embodiments, separation of the ADCs may be achieved by contacting an ADC mixture (e.g., a mixture comprising a drug loaded species of ADC of 4 or less and a drug loaded species of ADC of 6 or more) with a hydrophobic resin, wherein the amount of resin is sufficient to allow binding of the drug loaded species which is being removed from the ADC mixture. The resin and ADC mixture are mixed together, such that the ADC species being removed (e.g., a drug loaded species of 6 or more) binds to the resin and can be separated from the other ADC species in the ADC mixture. The amount of resin used in the method is based on a weight ratio between the species to be removed and the resin, where the amount of resin used does not allow for significant binding of the drug loaded species that is desired. Thus, methods may be used to reduce the average DAR to less than 4. Further, the purification methods described herein may be used to isolate ADCs having any desired range of drug loaded species, e.g., a drug loaded species of 4 or less, a drug loaded species of 3 or less, a drug loaded species of 2 or less, a drug loaded species of 1 or less.
Certain species of molecule(s) binds to a surface based on hydrophobic interactions between the species and a hydrophobic resin. In one embodiment, method of the invention refers to a purification process that relies upon the intermixing of a hydrophobic resin and a mixture of ADCs, wherein the amount of resin added to the mixture determines which species (e.g., ADCs with a DAR of 6 or more) will bind. Following production and purification of an antibody from an expression system (e.g., a mammalian expression system), the antibody is reduced and coupled to a drug through a conjugation reaction. The resulting ADC mixture often contains ADCs having a range of DARs, e.g., 1 to 8. In one embodiment, the ADC mixture comprises a drug loaded species of 4 or less and a drug loaded species of 6 or more. According to the methods of the invention, the ADC mixture may be purified using a process, such as, but not limited to, a batch process, such that ADCs having a drug loaded species of 4 or less are selected and separated from ADCs having a higher drug load (e.g., ADCs having a drug loaded species of 6 or more). Notably, the purification methods described herein may be used to isolate ADCs having any desired range of DAR, e.g., a DAR of 4 or less, a DAR of 3 or less, or a DAR of 2 or less.
Thus, in one embodiment, an ADC mixture comprising a drug loaded species of 4 or less and a drug loaded species of 6 or more may be contacted with a hydrophobic resin to form a resin mixture, wherein the amount of hydrophobic resin contacted with the ADC mixture is sufficient to allow binding of the drug loaded species of 6 or more to the resin but does not allow significant binding of the drug load species of 4 or less; and removing the hydrophobic resin from the ADC mixture, such that the composition comprising ADCs is obtained, wherein the composition comprises less than 15% of the drug loaded species of 6 or more, and wherein the ADC comprises an antibody conjugated to a Bcl-xL inhibitor. In a separate embodiment, the method of the invention comprises contacting an ADC mixture comprising a drug loaded species of 4 or less and a drug loaded species of 6 or more with a hydrophobic resin to form a resin mixture, wherein the amount of hydrophobic resin contacted with the ADC mixture is sufficient to allow binding of the drug loaded species of 6 or more to the resin but does not allow significant binding of the drug load species of 4 or less; and removing the hydrophobic resin from the ADC mixture, such that the composition comprising ADCs is obtained, wherein the composition comprises less than 15% of the drug loaded species of 6 or more, and wherein the ADC comprises an antibody conjugated to a Bcl-xL inhibitor, wherein the hydrophobic resin weight is 3 to 12 times the weight of the drug loaded species of 6 or more in the ADC mixture.
The ADC separation method described herein method may be performed using a batch purification method. The batch purification process generally includes adding the ADC mixture to the hydrophobic resin in a vessel, mixing, and subsequently separating the resin from the supernatant. For example, in the context of batch purification, a hydrophobic resin may be prepared in or equilibrated to the desired equilibration buffer. A slurry of the hydrophobic resin may thus be obtained. The ADC mixture may then be contacted with the slurry to adsorb the specific species of ADC(s) to be separated by the hydrophobic resin. The solution comprising the desired ADCs that do not bind to the hydrophobic resin material may then be separated from the slurry, e.g., by filtration or by allowing the slurry to settle and removing the supernatant. The resulting slurry can be subjected to one or more washing steps. In order to elute bound ADCs, the salt concentration can be decreased. In one embodiment, the process used in the invention includes no more than 50 g of hydrophobic resin.
Thus, a batch method may be used to contact an ADC mixture comprising a drug loaded species of 4 or less and a drug loaded species of 6 or more with a hydrophobic resin to form a resin mixture, wherein the amount of hydrophobic resin contacted with the ADC mixture is sufficient to allow binding of the drug loaded species of 6 or more to the resin but does not allow significant binding of the drug load species of 4 or less; and removing the hydrophobic resin from the ADC mixture, such that the composition comprising ADCs is obtained, wherein the composition comprises less than 15% of the drug loaded species of 6 or more, and wherein the ADC comprises an antibody conjugated to a Bcl-xL inhibitor In a separate embodiment a batch method is used to contact an ADC mixture comprising a drug loaded species of 4 or less and a drug loaded species of 6 or more with a hydrophobic resin to form a resin mixture, wherein the amount of hydrophobic resin contacted with the ADC mixture is sufficient to allow binding of the drug loaded species of 6 or more to the resin but does not allow significant binding of the drug load species of 4 or less; and removing the hydrophobic resin from the ADC mixture, such that the composition comprising ADCs is obtained, wherein the composition comprises less than 15% of the drug loaded species of 6 or more, and wherein the ADC comprises an antibody conjugated to a Bcl-xL inhibitor, wherein the hydrophobic resin weight is 3 to 12 times the weight of the drug loaded species of 6 or more in the ADC mixture.
Alternatively, in a separate embodiment, purification may be performed using a circulation process, whereby the resin is packed in a container and the ADC mixture is passed over the hydrophobic resin bed until the specific species of ADC(s) to be separated have been removed. The supernatant (containing the desired ADC species) is then pumped from the container and the resin bed may be subjected to washing steps.
A circulation process may be used to contact an ADC mixture comprising a drug loaded species of 4 or less and a drug loaded species of 6 or more with a hydrophobic resin to form a resin mixture, wherein the amount of hydrophobic resin contacted with the ADC mixture is sufficient to allow binding of the drug loaded species of 6 or more to the resin but does not allow significant binding of the drug load species of 4 or less; and removing the hydrophobic resin from the ADC mixture, such that the composition comprising ADCs is obtained, wherein the composition comprises less than 15% of the drug loaded species of 6 or more, and wherein the ADC comprises an antibody conjugated to a Bcl-xL inhibitor. In a separate embodiment, a circulation process is used to contact an ADC mixture comprising a drug loaded species of 4 or less and a drug loaded species of 6 or more with a hydrophobic resin to form a resin mixture, wherein the amount of hydrophobic resin contacted with the ADC mixture is sufficient to allow binding of the drug loaded species of 6 or more to the resin but does not allow significant binding of the drug load species of 4 or less; and removing the hydrophobic resin from the ADC mixture, such that the composition comprising ADCs is obtained, wherein the composition comprises less than 15% of the drug loaded species of 6 or more, and wherein the ADC comprises an antibody conjugated to a Bcl-xL inhibitor, wherein the hydrophobic resin weight is 3 to 12 times the weight of the drug loaded species of 6 or more in the ADC mixture.
Alternatively, a flow through process may be used to purify an ADC mixture to arrive at a composition comprising a majority of ADCs having a certain desired DAR. In a flow through process, resin is packed in a container, e.g., a column, and the ADC mixture is passed over the packed resin such that the desired ADC species does not substantially bind to the resin and flows through the resin, and the undesired ADC species is bound to the resin. A flow through process may be performed in a single pass mode (where the ADC species of interest are obtained as a result of a single pass through the resin of the container) or in a multi-pass mode (where the ADC species of interest are obtained as a result of multiple passes through the resin of the container) The flow through process is performed such that the weight of resin selected binds to the undesired ADC population, and the desired ADCs (e.g., DAR 2-4) flow over the resin and are collected in the flow through after one or multiple passes.
A flow through process may be used to contact an ADC mixture comprising a drug loaded species of 4 or less and a drug loaded species of 6 or more with a hydrophobic resin, wherein the amount of hydrophobic resin contacted with the ADC mixture is sufficient to allow binding of the drug loaded species of 6 or more to the resin but does not allow significant binding of the drug load species of 4 or less, where the drug load species of 4 or less passes over the resin and is subsequently collected after one or multiple passes, such that the composition comprising the desired ADCs (e.g. DAR 2-4) is obtained, wherein the composition comprises less than 15% of the drug loaded species of 6 or more, and wherein the ADC comprises an antibody conjugated to a Bcl-xL inhibitor. In a separate embodiment, a flow through process is used to contact an ADC mixture comprising a drug loaded species of 4 or less and a drug loaded species of 6 or more with a hydrophobic resin by passing the ADC mixture over the resin, wherein the amount of hydrophobic resin contacted with the ADC mixture is sufficient to allow binding of the drug loaded species of 6 or more to the resin but does not allow significant binding of the drug load species of 4 or less, where the drug load species of 4 or less passes over the resin and is subsequently collected, such that the composition comprising ADCs is obtained, wherein the composition comprises less than 15% of the drug loaded species of 6 or more, and wherein the ADC comprises an antibody conjugated to a a drug, e.g., a Bcl-xL inhibitor, wherein the amount of hydrophobic resin weight is 3 to 12 times the weight of the drug loaded species of 6 or more in the ADC mixture.
Following a flow through process, the resin may be washed with a one or more washes following in order to further recover ADCs having the desired DAR range (found in the wash filtrate). For example, a plurality of washes having decreasing conductivity may be used to further recover ADCs having the DAR of interest. The elution material obtained from the washing of the resin may be subsequently combined with the filtrate resulting from the flow through process for improved recovery of ADCs having the DAR of interest.
The aforementioned batch, circulation, and flow through process purification methods are based on the use of a hydrophobic resin to separate high vs. low drug loaded species of ADC.
Hydrophobic resin comprises hydrophobic groups which interact with the hydrophobic properties of the ADCs. Hydrophobic groups on the ADC interact with hydrophobic groups within the hydrophobic resin. The more hydrophobic a protein is the stronger it will interact with the hydrophobic resin.
Hydrophobic resin normally comprises a base matrix (e.g., cross-linked agarose or synthetic copolymer material) to which hydrophobic ligands (e.g., alkyl or aryl groups) are coupled. Many hydrophobic resins are available commercially. Examples include, but are not limited to, Phenyl SepharoseTM 6 Fast Flow with low or high substitution (Pharmacia LKB Biotechnology AB Sweden); Phenyl SepharoseTM High Performance (Pharmacia LKB Biotechnology, AB, Sweden); Octyl SepharoseTM High Performance (Pharmacia LKB Biotechnology, AB, Sweden); FractogelTM EMD Propyl or FractogelTM EMD Phenyl columns (E. Merck, Germany); Macro-PrepTM Methyl or Macro-PrepTM. t-Butyl Supports (Bio-Rad, California); WP HI-Propyl (C3)TM (J. T. Baker, New Jersey); and ToyopearlTM ether, hexyl, phenyl or butyl (TosoHaas, PA). In one embodiment, the hydrophobic resin is a butyl hydrophobic resin. In another embodiment, the hydrophobic resin is a phenyl hydrophobic resin. In another embodiment, the hydrophobic resin is a hexyl hydrophobic resin, an octyl hydrophobic resin, or a decyl hydrophobic resin. In one embodiment, the hydrophobic resin is a methacrylic polymer having n-butyl ligands (e.g. TOYOPEARL Butyl-600M).
Further methods for purifying ADC mixtures to obtain a composition having a desired DAR are described in U.S. Application No.14/210,602 (U.S. Patent Appln. Publication No. US
2014/0286968), incorporated by reference in its entirety.
In certain embodiments of the invention, ADCs described herein having a DAR2 are purified from ADCs having higher or lower DARs. Such purified DAR2 ADCs are referred to herein as“E2”. Purification methods for achieving a composition having E2 anti-B7-H3 ADCs. In one embodiment, of the invention provides a composition comprising an ADC mixture, wherein at least 75% of the ADCs are anti-B7H3 ADCs (like those described herein) having a DAR2. In another embodiment, the invention provides a composition comprising an ADC mixture, wherein at least 80% of the ADCs are anti-B7H3 ADCs (like those described herein) having a DAR2. In another embodiment, the invention provides a composition comprising an ADC mixture, wherein at least 85% of the ADCs are anti-B7H3 ADCs (like those described herein) having a DAR2. In another embodiment, the invention provides a composition comprising an ADC mixture, wherein at least 90% of the ADCs are anti- B7H3 ADCs (like those described herein) having a DAR2.
V. Uses of Anti-B7-H3 Antibodies and Anti-B7-H3 ADCs
The antibodies and ADCs of the invention preferably are capable of neutralizing human B7- H3 activity both in vivo. Accordingly, such antibodies and ADCs of the invention can be used to inhibit hB7-H3 activity, e.g., in a cell culture containing hB7-H3, in human subjects or in other mammalian subjects having B7-H3 with which an antibody of the invention cross-reacts. In one embodiment, the invention provides a method for inhibiting hB7-H3 activity comprising contacting hB7-H3 with an antibody or ADC of the invention such that hB7-H3 activity is inhibited. For example, in a cell culture containing, or suspected of containing hB7-H3, an antibody or antibody portion of the invention can be added to the culture medium to inhibit hB7-H3 activity in the culture.
In another embodiment, of the invention a method for reducing hB7-H3 activity in a subject, advantageously from a subject suffering from a disease or disorder in which B7-H3 activity is detrimental The invention provides methods for reducing B7-H3 activity in a subject suffering from such a disease or disorder, which method comprises administering to the subject an antibody or ADC of the invention such that B7-H3 activity in the subject is reduced. Preferably, the B7-H3 is human B7-H3, and the subject is a human subject. Alternatively, the subject can be a mammal expressing a B7-H3 to which antibodies of the invention are capable of binding. Still further the subject can be a mammal into which B7-H3 has been introduced (e.g., by administration of B7-H3 or by expression of a B7-H3 transgene). Antibodies or ADCs of the invention can be administered to a human subject for therapeutic purposes. Moreover, antibodies or ADCS of the invention can be administered to a non- human mammal expressing a B7-H3 with which the antibody is capable of binding for veterinary purposes or as an animal model of human disease. Regarding the latter, such animal models may be useful for evaluating the therapeutic efficacy of antibodies of the invention (e.g., testing of dosages and time courses of administration).
As used herein, the term“a disorder in which B7-H3 expression is detrimental” is intended to include diseases and other disorders in which the presence of B7-H3 in a subject suffering from the disorder has been shown to be expressed, or has been shown to be or is suspected of being either responsible for the pathophysiology of the disorder or a factor that contributes to the disorder. For example, the ADCs of the invention may be used to target tumor cells that are expressing B7-H3. Non-limiting examples of disorders that can be treated with the antibodies or ADCs of the invention, for example, an ADC comprising huAb13v1, huAb13v1, or antigen binding fragments thereof, include,, but are not limited to, a variety of cancers including, but not limited to, small cell lung cancer, non small cell lunch cancer (NSCLC), breast cancer, ovarian cancer, lung cancer, a glioma, prostate cancer, pancreatic cancer, colon cancer, head and neck cancer, leukemia, e.g., acute myeloid leukemia (AML), lymphoma, e.g., non-Hodgkin's lymphoma (NHL), and kidney cancer. Other examples of cancer that may be treated using the compositions and methods disclosed herein include squamous cell carcinoma (e.g., squamous lung cancer or squamous head and neck cancer), triple negative breast cancer, non-small cell lung cancer, colorectal cancer, and mesothelioma. In one embodiment, the antibodies and ADCs disclosed herein are used to treat a solid tumor, e.g., inhibit growth of or decrease size of a solid tumor, overexpressing B7-H3 or which is B7-H3 positive. In one embodiment, the invention is directed to the treatment of squamous lung cancer associated with B7- H3 expression. In another embodiment, the antibodies and ADCs disclosed herein are used to treat triple negative breast cancer (TNBC). Diseases and disorders described herein may be treated by anti- B7-H3 antibodies or ADCs of the invention, as well as pharmaceutical compositions comprising such anti-B7-H3 antibodies or ADCs.
In certain embodiments, the cancer may be characterized as having EGFR overexpression. In one embodiment, the ADCs of the invention may be used to treating cancer associated with an activating EGFR mutation. Examples of such mutations include, but are not limited to, an exon 19 deletion mutation, a single-point substitution mutation L858R in exon 21, a T790M point mutation, and combinations thereof In certain embodiments, the antibodies and ADCs disclosed herein are administered to a subject in need thereof in order to treat advanced solid tumor types likely to exhibit elevated levels of B7-H3. Examples of such tumors include, but are not limited to, small cell lung cancer, breast cancer, ovarian cancer, head and neck squamous cell carcinoma, non-small cell lung cancer, triple negative breast cancer, colorectal carcinoma, and glioblastoma multiforme.
In certain embodiments, the invention includes a method for inhibiting or decreasing solid tumor growth in a subject having a solid tumor, said method comprising administering an anti-B7-H3 antibody or ADC described herein, to the subject having the solid tumor, such that the solid tumor growth is inhibited or decreased. In certain embodiments, the solid tumor is a non-small cell lung carcinoma or a glioblastoma. In further embodiments, the solid tumor is a B7-H3-expressing solid tumors. In further embodiments, the solid tumor is an B7-H3 overexpressing solid tumors. In certain embodiments the anti-B7-H3 antibodies or ADCs described herein are are administered to a subject having glioblastoma multiforme, alone or in combination with an additional agent, e.g., radiation and/or temozolomide.
In certain embodiments the anti-B7-H3 ADCs described herein are are administered to a subject having small cell lung cancer, alone or in combination with an additional agent, e.g., ABT-199 (venetoclax).
In certain embodiments the anti-B7-H3 ADCs described herein are administered to a subject having non-small cell lung cancer, alone or in combination with an additional agent, e.g., a taxane. In certain embodiments the anti-B7-H3 antibodies or ADCs described herein are administered to a subject having breast cancer, alone or in combination with an additional agent, e.g., a taxane. In certain embodiments the anti-B7-H3 antibodies or ADCs described herein are administered to a subject having ovarian cancer, alone or in combination with an additional agent, e.g., a taxane.
Other combination therapies which are included in the invention are the administration of an anti-B7-H3 ADC with an agent selected from the group consisting of an anti-PD1 antibody (e.g. pembrolizumab), an anti-PD-L1 antibody (e.g. atezolizumab), an anti-CTLA-4 antibody (e.g.
ipilimumab), a MEK inhibitor (e.g. trametinib), an ERK inhibitor, a BRAF inhibitor (e.g. dabrafenib), osimertinib, erlotinib, gefitinib, sorafenib, a CDK9 inhibitor (e.g. dinaciclib), a MCL-1 inhibitor, temozolomide, a Bcl-xL inhibitor, a Bcl-2 inhibitor (e.g. venetoclax), ibrutinib, a mTOR inhibitor (e.g. everolimus), a PI3K inhibitor (e.g. buparlisib), duvelisib, idelalisib, an AKT inhibitor, a HER2 inhibitor (e.g. lapatinib), a taxane (e.g. docetaxel, paclitaxel, nab-paclitaxel), an ADC comprising an auristatin, an ADC comprising a PBD (e.g. rovalpituzumab tesirine), an ADC comprising a maytansinoid (e.g. TDM1), a TRAIL agonist, a proteasome inhibitor (e.g. bortezomib), and a nicotinamide phosphoribosyltransferase (NAMPT) inhibitor.
Combination therapies include administration of an ADC of the invention prior to, concurrently with, or following administration of an additional therapeutic agent, including those described above In certain embodiments, the invention includes a method for inhibiting or decreasing solid tumor growth in a subject having a solid tumor which was identified as an B7-H3 expressing or B7- H3 overexpressing tumor, said method comprising administering an anti-B7-H3 antibody or ADC described herein, to the subject having the solid tumor, such that the solid tumor growth is inhibited or decreased. Methods for identifying B7-H3 expressing tumors (e.g., B7-H3 overexpressing tumors) are known in the art, and include FDA-approved tests and validation assays. For example, the B7-H3 assay is a qualitative immunohistochemical (IHC) kit system used to identify B7-H3 expression in normal and neoplastic tissues routinely-fixed for histological evaluation. In addition, PCR-based assays may also be used for identifying B7-H3 overexpressing tumors. The amplified PCR products may be subsequently analyzed, for example, by gel electrophoresis using standard methods known in the art to determine the size of the PCR products. Such tests may be used to identify tumors that may be treated with the methods and compositions described herein.
Any of the methods for gene therapy available in the art can be used according to the invention. For general reviews of the methods of gene therapy, see Goldspiel et al., 1993, Clinical Pharmacy 12:488-505; Wu and Wu, 1991, Biotherapy 3:87-95; Tolstoshev, 1993, Ann. Rev.
Pharmacol. Toxicol.32:573-596; Mulligan, Science 260:926- 932 (1993); and Morgan and Anderson, 1993, Ann. Rev. Biochem.62:191-217; May, 1993, TIBTECH 11(5):155-215. Methods commonly known in the art of recombinant DNA technology which can be used are described in Ausubel et al. (eds.), Current Protocols in Molecular Biology, John Wiley &Sons, NY (1993); and Kriegler, Gene Transfer and Expression, A Laboratory Manual, Stockton Press, NY (1990). Detailed description of various methods of gene therapy is provided in US20050042664 A1 which is incorporated herein by reference.
In another aspect, this application features a method of treating (e.g., curing, suppressing, ameliorating, delaying or preventing the onset of, or preventing recurrence or relapse of) or preventing a B7-H3-associated disorder, in a subject. The method includes: administering to the subject an B7- H3 binding agent, e.g., an anti-B7-H3 antibody or fragment thereof as described herein, in an amount sufficient to treat or prevent the B7-H3-associated disorder. The B7-H3 antagonist, e.g., the anti-B7- H3 antibody or fragment thereof, can be administered to the subject, alone or in combination with other therapeutic modalities as described herein.
Antibodies or ADCs of the invention, or antigen binding portions thereof can be used alone or in combination to treat such diseases. It should be understood that the antibodies of the invention or antigen binding portion thereof can be used alone or in combination with an additional agent, e.g., a therapeutic agent, said additional agent being selected by the skilled artisan for its intended purpose. For example, the additional agent can be a therapeutic agent art-recognized as being useful to treat the disease or condition being treated by the antibody of the invention. The additional agent also can be an agent that imparts a beneficial attribute to the therapeutic composition, e.g., an agent which affects the viscosity of the composition It should further be understood that the combinations which are to be included within this invention are those combinations useful for their intended purpose. The agents set forth below are illustrative for purposes and not intended to be limited. The combinations, which are part of this invention, can be the antibodies of the invention and at least one additional agent selected from the lists below. The combination can also include more than one additional agent, e.g., two or three additional agents if the combination is such that the formed composition can perform its intended function.
The combination therapy can include one or more B7-H3 antagonists, e.g., anti-B7-H3 antibodies or fragments thereof, formulated with, and/or co-administered with, one or more additional therapeutic agents, e.g., one or more cytokine and growth factor inhibitors, immunosuppressants, anti- inflammatory agents (e.g., systemic anti-inflammatory agents), anti-fibrotic agents, metabolic inhibitors, enzyme inhibitors, and/or cytotoxic or cytostatic agents, mitotic inhibitors, antitumor antibiotics, immunomodulating agents, vectors for gene therapy, alkylating agents, antiangiogenic agents, antimetabolites, boron-containing agents, chemoprotective agents, hormones, antihormone agents, corticosteroids, photoactive therapeutic agents, oligonucleotides, radionuclide agents, topoisomerase inhibitors, kinase inhibitors, or radiosensitizers, as described in more herein.
In a particular embodiment, the anti-B7-H3 binding proteins described herein, for example, anti-B7-H3 antibodies, are used in combination with an anti-cancer agent or an antineoplastic agent. The terms“anti-cancer agent” and“antineoplastic agent” refer to drugs used to treat malignancies, such as cancerous growths. Drug therapy may be used alone, or in combination with other treatments such as surgery or radiation therapy. Several classes of drugs may be used in cancer treatment, depending on the nature of the organ involved. For example, breast cancers are commonly stimulated by estrogens, and may be treated with drugs which inactive the sex hormones. Similarly, prostate cancer may be treated with drugs that inactivate androgens, the male sex hormone. Anti-cancer agents that may be used in conjunction with the anti-B7-H3 antibodies or ADCs of the invention include, among others, an anti-PD1 antibody (e.g., pembrolizumab), an anti-PD-L1 antibody (e.g. atezolizumab), an anti-CTLA-4 antibody (e.g., ipilimumab), a MEK inhibitor (e.g., trametinib), an ERK inhibitor, a BRAF inhibitor (e.g., dabrafenib), osimertinib (AZD9291), erlotinib, gefitinib, sorafenib, a CDK9 inhibitor (e.g., dinaciclib), a MCL-1 inhibitor, temozolomide, a Bcl-xL inhibitor, a Bcl-2 inhibitor (e.g. venetoclax), ibrutinib, a mTOR inhibitor (e.g., everolimus), a PI3K inhibitor (e.g., buparlisib), duvelisib, idelalisib, an AKT inhibitor, a HER2 inhibitor (e.g., lapatinib), Herceptin, a taxane (e.g. docetaxel, paclitaxel, nab-paclitaxel), an ADC comprising an auristatin, an ADC comprising a PBD (e.g., rovalpituzumab tesirine), an ADC comprising a maytansinoid (e.g., TDM1), a TRAIL agonist, a proteasome inhibitor (e.g., bortezomib), and a nicotinamide
phosphoribosyltransferase (NAMPT) inhibitor, as well as the following agents: Anti-Cancer Agent Comments Examples Antibodies Antibodies which bind IGF- A12 (fully humanized mAb)
1R (insulin-like growth 19D12 (fully humanized mAb)
factor type 1 receptor), Cp751-871 (fully humanized mAb) which is expressed on the H7C10 (humanized mAb)
cell surface of most human alphaIR3 (mouse)
cancers ScFV/FC (mouse/human chimera)
EM/164 (mouse)
Antibodies which bind
EGFR; Mutations affecting Matuzumab (EMD72000)
EGFR expression or activity Erbitux® / Cetuximab (Imclone) could result in cancer Vectibix® / Panitumumab (Amgen) _____________________ mAb 806
Antibodies which bind Nimotuxumab (TheraCIM)
cMET (Mesechymal
epithelial transition factor); AVEO (AV299) (AVEO)
a member of the MET AMG102 (Amgen) family of receptor tyrosine 5D5 (OA-5d5) (Genentech)
kinases) H244G11 (Pierre Fabre)
Anti-ErbB3 antibodies Ab #14 (MM 121-14)
Herceptin® (Trastuzumab; Genentech) 1B4C3; 2D1D12 (U3 Pharma AG)
Small Molecules Insulin-like growth factor NVP-AEW541-A
Targeting IGF1R type 1 receptor which is BMS-536,924 (1H-benzoimidazol-2-yl)-1H- expressed on the cell pyridin-2-one)
surface of many human BMS-554,417
cancers Cycloligan
TAE226
PQ401
Small Molecules EGFR; Iressa® / Gefitinib (AstraZeneca)
Targeting EGFR Overexpression or CI-1033 (PD 183805) (Pfizer)
mutations affecting EGFR Lapatinib (GW-572016) (GlaxoSmithKline) expression or activity could Tykerb® / Lapatinib Ditosylate (Smith Kline result in cancer Beecham)
Tarceva ® / Erlotinib HCL (OSI-774) (OSI Pharma)
PKI-166 (Novartis)
PD-158780
EKB-569
Tyrphostin AG 1478 (4-(3-Chloroanillino)- 6,7-dimethoxyquinazoline)
Small Molecules cMET (Mesenchymal PHA665752
Targeting cMET epithelial transition factor); ARQ 197
a member of the MET
family of receptor tyrosine
kinases)
Antimetabolites Flourouracil (5-FU) Capecitabine / XELODA® (HLR Roche)
5-Trifluoromethyl-2’-deoxyuridine
Methotrexate sodium (Trexall) (Barr) Raltitrexed/ Tomudex® (AstraZeneca) Pemetrexed / Alimta® (Lilly)
Tegafur
Cytosine Arabinoside (Cytarabine, Ara-C) / Thioguanine® (GlaxoSmithKline)
5-azacytidine
6-mercaptopurine (Mercaptopurine, 6-MP) Azathioprine / Azasan® (AAIPHARMA LLC) 6-thioguanine (6-TG) / Purinethol® (TEVA) Pentostatin / Nipent® (Hospira Inc.)
Fludarabine phosphate / Fludara® (Bayer Health Care)
Cladribine (2-CdA, 2-chlorodeoxyadenosine) / Leustatin® (Ortho Biotech)
Alkylating agents An alkylating antineoplastic Ribonucleotide Reductase Inhibitor (RNR) agent is an alkylating agent Cyclophosphamide / Cytoxan (BMS) that attaches an alkyl group Neosar (TEVA)
to DNA. Since cancer cells Ifosfamide / Mitoxana® (ASTA Medica) generally proliferate Thiotepa (Bedford, Abraxis, Teva) unrestrictively more than do BCNU→ 1,3-bis(2-chloroethyl)-1-nitosourea healthy cells they are more CCNU→ 1, -(2-chloroethyl)-3-cyclohexyl-1- sensitive to DNA damage, nitrosourea (methyl CCNU)
and alkylating agents are Hexamethylmelamine (Altretamine, HMM) / used clinically to treat a Hexalen® (MGI Pharma Inc.)
variety of tumors. Busulfan / Myleran (GlaxoSmithKline)
Procarbazine HCL/ Matulane (Sigma Tau Pharmaceuticals, Inc.)
Dacarbazine (DTIC)
Chlorambucil / Leukara® (SmithKline Beecham)
Melphalan / Alkeran® (GlaxoSmithKline) Cisplatin (Cisplatinum, CDDP) / Platinol (Bristol Myers)
Carboplatin / Paraplatin (BMS)
Oxaliplatin /Eloxitan® (Sanofi-Aventis US) Topoisomerase Topoisomerase inhibitors Doxorubicin HCL / Doxil® (Alza) inhibitors are chemotherapy agents Daunorubicin citrate / Daunoxome® (Gilead) designed to interfere with Mitoxantrone HCL / Novantrone (EMD the action of topoisomerase Serono)
enzymes (topoisomerase I Actinomycin D
and II), which are enzymes Etoposide / Vepesid® (BMS)/ Etopophos® that control the changes in (Hospira, Bedford, Teva Parenteral, Etc.) DNA structure by Topotecan HCL / Hycamtin®
catalyzing the breaking and (GlaxoSmithKline)
rejoining of the Teniposide (VM-26) / Vumon® (BMS) phosphodiester backbone of Irinotecan HCL(CPT-ll) / Camptosar® DNA strands during the (Pharmacia & Upjohn) normal cell cycle.
Microtubule Microtubules are one of the Vincristine / Oncovin® (Lilly) targeting agents components of the Vinblastine sulfate / Velban®(discontinued) cytoskeleton. They have (Lilly)
diameter of ~24 nm and Vinorelbine tartrate / Navelbine®
length varying from several (PierreFabre)
micrometers to possibly Vindesine sulphate / Eldisine® (Lilly) millimeters in axons of Pac1itaxel / Taxol® (BMS)
nerve cells. Microtubules Docetaxel / Taxotere® (Sanofi Aventis US) serve as structural Nanoparticle paclitaxel (ABI-007) / components within cells and Abraxane® (Abraxis BioScience, Inc.) are involved in many Ixabepilone / IXEMPRA™ (BMS) cellular processes including
mitosis, cytokinesis, and
vesicular transport.
Kinase inhibitors Kinases are enzymes that Imatinib mesylate / Gleevec (Novartis)
catalyzes the transfer of Sunitinib malate / Sutent® (Pfizer) phosphate groups from Sorafenib tosylate / Nexavar® (Bayer) high-energy, phosphate- Nilotinib hydrochloride monohydrate / donating molecules to Tasigna® (Novartis), Osimertinib, specific substrates, and are Cobimetinib, Trametinib, Dabrafenib, utilized to transmit signals Dinaciclib
and regulate complex
processes in cells.
Protein synthesis Induces cell apoptosis L-asparaginase / Elspar® (Merck & Co.) inhibitors
Immunotherapeutic Induces cancer patients to Alpha interferon
agents exhibit immune Angiogenesis Inhibitor / Avastin®
responsiveness (Genentech)
IL-2→ Interleukin 2 (Aldesleukin) / Proleukin ® (Chiron)
IL-12→ Interleukin 12
Antibody / small molecule
immune checkpoint Anti-CTLA-4 and PR-1 therapies
modulators Yervoy® (ipilimumab; Bristol-Myers Squibb)
Opdivo® (nivolumab; Bristol-Myers Squibb) Keytrada® (pembrolizumab; Merck)
Hormones Hormone therapies Toremifene citrate / Fareston® (GTX, Inc.) associated with menopause Fulvestrant / Faslodex® (AstraZeneca) and aging seek to increase Raloxifene HCL / Evista® (Lilly)
the amount of certain Anastrazole / Arimidex® (AstraZeneca) hormones in your body to Letrozole / Femara® (Novartis)
compensate for age- or Fadrozole (CGS 16949A )
disease-related hormonal Exemestane / Aromasin® (Pharmacia & declines. Hormone therapy Upjohn)
as a cancer treatment either Leuprolide acetate / Eligard® (QTL USA) reduces the level of specific Lupron® (TAP Pharm) hormones or alters the Goserelin acetate / Zoladex® (AstraZeneca) cancer’s ability to use these Triptorelin pamoate / Trelstar® (Watson Labs) hormones to grow and Buserelin / Suprefact® (Sanofi Aventis) spread. Nafarelin / Synarel® (Pfizer)
Cetrorelix / Cetrotide® (EMD Serono) Bicalutamide / Casodex® (AstraZeneca) Nilutamide / Nilandron® (Aventis Pharm ) Megestrol acetate / Megace® (BMS)
Somatostatin Analogs (Octreotide acetate / Sandostatin® (Novartis)
Glucocorticoids Anti-inflammatory drugs Prednisolone
used to reduce swelling that Dexamethasone / Decadron® (Wyeth) causes cancer pain.
Aromatose inhibitors Includes imidazoles Ketoconazole
mTOR inhibitors the mTOR signaling Sirolimus (Rapamycin) / Rapamune® (Wyeth) pathway was originally Temsirolimus (CCI-779) / Torisel® (Wyeth) discovered during studies of Deforolimus (AP23573) / (Ariad Pharm.) the immunosuppressive Everolimus (RAD00I) / Certican® (Novartis) agent rapamycin. This
highly conserved pathway
regulates cell proliferation
and metabolism in response
to environmental factors,
linking cell growth factor
receptor signaling via
phosphoinositide-3- kinase(PI-3K) to cell
growth, proliferation, and
angiogenesis.
In addition to the above anti-cancer agents, the anti-B7-H3 antibodies and ADCs described herein may be administered in combination with the agents described herein. Further, the aforementioned anti-cancer agents may also be used in the ADCs of the invention.
In particular embodiments, the anti-B7-H3 antibodies or ADCs can be administered alone or with another anti-cancer agent which acts in conjunction with or synergistically with the antibody to treat the disease associated with B7-H3 activity. Such anti-cancer agents include, for example, agents well known in the art (e.g., cytotoxins, chemotherapeutic agents, small molecules and radiation). Examples of anti-cancer agents include, but are not limited to, Panorex (Glaxo-Welcome), Rituxan (IDEC/Genentech/Hoffman la Roche), Mylotarg (Wyeth), Campath (Millennium), Zevalin (IDEC and Schering AG), Bexxar (Corixa/GSK), Erbitux (Imclone/BMS), Avastin (Genentech) and Herceptin (Genentech/Hoffman la Roche). Other anti-cancer agents include, but are not limited to, those disclosed in U.S. Patent No.7,598,028 and International Publication No. WO2008/100624, the contents of which are hereby incorporated by reference. One or more anti-cancer agents may be administered either simultaneously or before or after administration of an antibody or antigen binding portion thereof of the invention.
In particular embodiments of the invention, the anti-B7-H3 antibodies or ADCs described herein can be used in a combination therapy with an apoptotic agent, such as a Bcl-xL inhibitor or a Bcl-2 (B-cell lymphoma 2) inhibitor (e.g., ABT-199 (venetoclax)) to treat cancer, such as leukemia, in a subject. In one embodiment, the anti-B7-H3 antibodies or ADCs described herein can be used in a combination therapy with a Bcl-xL inhibitor for treating cancer. In one embodiment, the anti-B7-H3 antibodies or ADCs described herein can be used in a combination therapy with venetoclax for treating cancer.
In particular embodiments of the invention, the anti-B7-H3 antibodies or ADCs described herein can be used in a combination therapy with an inhibitor of NAMPT (see examples of inhibitors in US 2013/0303509; AbbVie, Inc., incorporated by reference herein) to treat a subject in need thereof. NAMPT (also known as pre-B-cell-colony-enhancing factor (PBEF) and visfatin) is an enzyme that catalyzes the phosphoribosylation of nicotinamide and is the rate-limiting enzyme in one of two pathways that salvage NAD. In one embodiment of the invention, anti-B7-H3 antibodies and ADCs described herein are administered in combination with a NAMPT inhibitor for the treatment of cancer in a subject.
In particular embodiments of the invention, the anti-B7-H3 antibodies or ADCs described herein can be used in a combination therapy with SN-38, which is the active metabolite of the topoisomerase inhibitor irinotecan.
In other embodiments of the invention, the anti-B7-H3 antibodies or ADCs described herein can be used in a combination therapy with a PARP (poly ADP ribose polymerase) inhibitor, e.g.,veliparib, to treat cancer, including breast, ovarian and non-small cell lung cancers.
Further examples of additional therapeutic agents that can be co-administered and/or formulated with anti-B7-H3 antibodies or anti-B7-H3 ADCs described herein, include, but are not limited to, one or more of: inhaled steroids; beta-agonists, e.g., short-acting or long- acting beta- agonists; antagonists of leukotrienes or leukotriene receptors; combination drugs such as ADVAIR; IgE inhibitors, e.g., anti-IgE antibodies (e.g., XOLAIR®, omalizumab); phosphodiesterase inhibitors (e.g., PDE4 inhibitors); xanthines; anticholinergic drugs; mast cell-stabilizing agents such as cromolyn; IL-4 inhibitors; IL-5 inhibitors; eotaxin/CCR3 inhibitors; antagonists of histamine or its receptors including H1, H2, H3, and H4, and antagonists of prostaglandin D or its receptors (DP1 and CRTH2). Such combinations can be used to treat, for example, asthma and other respiratory disorders. Other examples of additional therapeutic agents that can be co-administered and/or formulated with anti-B7-H3 antibodies or anti-B7-H3 ADCs described herein, include, but are not limited to, one or more of, an anti-PD1 antibody (e.g., pembrolizumab), an anti-PD-L1 antibody (e.g. atezolizumab), an anti-CTLA-4 antibody (e.g., ipilimumab), a MEK inhibitor (e.g., trametinib), an ERK inhibitor, a BRAF inhibitor (e.g., dabrafenib), osimertinib (AZD9291), erlotinib, gefitinib, sorafenib, a CDK9 inhibitor (e.g., dinaciclib), a MCL-1 inhibitor, temozolomide, a Bcl-xL inhibitor, a Bcl-2 inhibitor (e.g. venetoclax), ibrutinib, a mTOR inhibitor (e.g., everolimus), a PI3K inhibitor (e.g., buparlisib), duvelisib, idelalisib, an AKT inhibitor, a HER2 inhibitor (e.g., lapatinib), Herceptin, a taxane (e.g. docetaxel, paclitaxel, nab-paclitaxel), an ADC comprising an auristatin, an ADC comprising a PBD (e.g., rovalpituzumab tesirine), an ADC comprising a maytansinoid (e.g., TDM1), a TRAIL agonist, a proteasome inhibitor (e.g., bortezomib), and a nicotinamide
phosphoribosyltransferase (NAMPT) inhibitor Additional examples of therapeutic agents that can be co-administered and/or formulated with one or more anti-B7-H3 antibodies or fragments thereof include one or more of: TNF antagonists (e.g., a soluble fragment of a TNF receptor, e.g., p55 or p75 human TNF receptor or derivatives thereof, e.g., 75 kD TNFR-IgG (75 kD TNF receptor-IgG fusion protein, ENBREL)); TNF enzyme antagonists, e.g., TNF converting enzyme (TACE) inhibitors; muscarinic receptor antagonists; TGF-beta antagonists; interferon gamma; perfenidone;
chemotherapeutic agents, e.g., methotrexate, leflunomide, or a sirolimus (rapamycin) or an analog thereof, e.g., CCI-779; COX2 and cPLA2 inhibitors; NSAIDs; immunomodulators; p38 inhibitors, TPL-2, MK-2 and NFkB inhibitors, among others.
Other preferred combinations are cytokine suppressive anti-inflammatory drug(s) (CSAIDs); antibodies to or antagonists of other human cytokines or growth factors, for example, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-15, IL-16, IL-18, IL-21, IL-31, interferons, EMAP-II, GM-CSF, FGF, EGF, PDGF, and edothelin-1, as well as the receptors of these cytokines and growth factors.
Antibodies of the invention, or antigen binding portions thereof, can be combined with antibodies to cell surface molecules such as CD2, CD3, CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD80 (B7.1), CD86 (B7.2), CD90, CTLA, CTLA-4, PD-1, or their ligands including CD154 (gp39 or CD40L).
Preferred combinations of therapeutic agents may interfere at different points in the inflammatory cascade; preferred examples include TNF antagonists like chimeric, humanized or human TNF antibodies, adalimumab, (HUMIRA; D2E7; PCT Publication No. WO 97/29131 and U.S. Patent No.6,090,382, incorporated by reference herein), CA2 (RemicadeTM), CDP 571, and soluble p55 or p75 TNF receptors, derivatives, thereof, (p75TNFR1gG (EnbrelTM) or p55TNFR1gG
(Lenercept), and also TNF converting enzyme (TACE) inhibitors; similarly IL-1 inhibitors
(Interleukin-1-converting enzyme inhibitors, IL-1RA etc.) may be effective for the same reason. Other preferred combinations include Interleukin 4.
The pharmaceutical compositions of the invention may include a“therapeutically effective amount” or a“prophylactically effective amount” of an antibody or antibody portion of the invention. A“therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result. A therapeutically effective amount of the antibody or antibody portion may be determined by a person skilled in the art and may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody or antibody portion to elicit a desired response in the individual. A therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody, or antibody portion, are outweighed by the therapeutically beneficial effects. A“prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount. Dosage regimens may be adjusted to provide the optimum desired response (e.g., a therapeutic or prophylactic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active compound and the particular therapeutic or prophylactic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals.
An exemplary, non-limiting range for a therapeutically or prophylactically effective amount of an ADC, an antibody or antibody portion of the invention is 0.1-20 mg/kg, more preferably 1-10 mg/kg. In one embodiment, the dose of the antibody or ADC described herein is 1 to 6 mg/kg, including the individual doses recited therein, e.g., 1 mg/kg, 2 mg/kg, 3 mg/kg, 4 mg/kg, 5 mg/kg, and 6 mg/kg. In another embodiment, the dose of the antibody or ADC described herein is 1 to 200 µg/kg, including the individual doses recited therein, e.g., 1 µg/kg, 2 µg/kg, 3 µg/kg, 4 µg/kg, 5 µg/kg, 10 µg/kg, 20 µg/kg, 30 µg/kg, 40 µg/kg, 50 µg/kg, 60 µg/kg, 80 µg/kg, 100 µg/kg, 120 µg/kg, 140 µg/kg, 160 µg/kg, 180 µg/kg and 200 µg/kg. It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition.
In one embodiment, an anti-B7-H3 ADC, including an ADC comprising antibody huAb13v1, huAb3v2.5, or huAb3v2.6, is administered to a subject in need thereof, e.g., a subject having cancer, at a dose of 0.1 to 30 mg/kg. In another embodiment, the anti-B7-H3 antibody, e.g., huAb13v1, huAb3v2.5, huAb3v2.6, or an antigen binding portion thereof, is administered to a subject in need thereof, e.g., a subject having cancer, as an ADC at a dose of 1 to 15 mg/kg. In another embodiment, the anti-B7-H3 antibody, e.g., huAb13v1, huAb3v2.5, huAb3v2.6, or an antigen binding portion thereof, is administered to a subject in need thereof, e.g., a subject having cancer, as an ADC at a dose of 1 to 10 mg/kg. In another embodiment, the anti-B7-H3 antibody, e.g., huAb13v1, huAb3v2.5, huAb3v2.6, or an antigen binding portion thereof, is administered to a subject in need thereof, e.g., a subject having cancer, as an ADC at a dose of 2 to 3. In another embodiment, the anti-B7-H3 antibody, e.g., huAb13v1, huAb3v2.5, huAb3v2.6, or an antigen binding portion thereof, is administered to a subject in need thereof, e.g., a subject having cancer, as an ADC at a dose of 1 to 4 mg/kg.
In one embodiment, an anti-B7-H3 antibody or ADC described herein, e.g., huAb13v1, huAb3v2.5, huAb3v2.6, is administered to a subject in need thereof, e.g., a subject having cancer, as an ADC at a dose of 1 to 200 µg/kg. In another embodiment, the anti-B7-H3 antibody, e.g., huAb13v1, huAb3v2.5, huAb3v2.6, or an antigen binding portion thereof, is administered to a subject in need thereof, e.g., a subject having cancer, as an ADC at a dose of 5 to 150 µg/kg. In another embodiment, the anti-B7-H3 antibody, e.g., huAb13v1, huAb3v2.5, huAb3v2.6, or an antigen binding portion thereof, is administered to a subject in need thereof, e.g., a subject having cancer, as an ADC at a dose of 5 to 100 µg/kg. In another embodiment, the anti-B7-H3 antibody, e.g., huAb13v1, huAb3v2.5, huAb3v2.6, or an antigen binding portion thereof, is administered to a subject in need thereof, e.g., a subject having cancer, as an ADC at a dose of 5 to 90 µg/kg. In another embodiment, the anti-B7-H3 antibody, e.g., huAb13v1, huAb3v2.5, huAb3v2.6, or an antigen binding portion thereof, is administered to a subject in need thereof, e.g., a subject having cancer, as an ADC at a dose of 5 to 80 µg/kg. In another embodiment, the anti-B7-H3 antibody, e.g., huAb13v1, huAb3v2.5, huAb3v2.6, or an antigen binding portion thereof, is administered to a subject in need thereof, e.g., a subject having cancer, as an ADC at a dose of 5 to 70 µg/kg. In another embodiment, the anti-B7-H3 antibody, e.g., huAb13v1, huAb3v2.5, huAb3v2.6, or an antigen binding portion thereof, is administered to a subject in need thereof, e.g., a subject having cancer, as an ADC at a dose of 5 to 60 µg/kg. In another embodiment, the anti-B7-H3 antibody, e.g., huAb13v1, huAb3v2.5, huAb3v2.6, or an antigen binding portion thereof, is administered to a subject in need thereof, e.g., a subject having cancer, as an ADC at a dose of 10 to 80 µg/kg.
Doses described above may be useful for the administration of either anti-B7-H3 ADCs or antibodies disclosed herein.
In another aspect, this application provides a method for detecting the presence of B7-H3 in a sample in vitro (e.g., a biological sample, such as serum, plasma, tissue, and biopsy). The subject method can be used to diagnose a disorder, e.g., a cancer. The method includes: (i) contacting the sample or a control sample with the anti-B7-H3 antibody or fragment thereof as described herein; and (ii) detecting formation of a complex between the anti-B7-H3 antibody or fragment thereof, and the sample or the control sample, wherein a statistically significant change in the formation of the complex in the sample relative to the control sample is indicative of the presence of B7-H3 in the sample.
Given their ability to bind to human B7-H3, the anti-human B7-H3 antibodies, or portions thereof, of the invention, (as well as ADCs thereof) can be used to detect human B7-H3 (e.g., in a biological sample, such as serum or plasma), using a conventional immunoassay, such as an enzyme linked immunosorbent assays (ELISA), an radioimmunoassay (RIA) or tissue immunohistochemistry. In one aspect the invention provides a method for detecting human B7-H3 in a biological sample comprising contacting a biological sample with an antibody, or antibody portion, of the invention and detecting either the antibody (or antibody portion) bound to human B7-H3 or unbound antibody (or antibody portion), to thereby detect human B7-H3 in the biological sample. The antibody is directly or indirectly labeled with a detectable substance to facilitate detection of the bound or unbound antibody. Suitable detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, β-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; and examples of suitable radioactive material include 3H 14
, C 35
, S, 90Y, 99Tc, 111In, 125I, 131I, 177Lu, 166Ho, or 153Sm.
Alternative to labeling the antibody, human B7-H3 can be assayed in biological fluids by a competition immunoassay utilizing rhB7-H3 standards labeled with a detectable substance and an unlabeled anti-human B7-H3 antibody. In this assay, the biological sample, the labeled rhB7-H3 standards and the anti-human B7-H3 antibody are combined and the amount of labeled rhB7-H3 standard bound to the unlabeled antibody is determined. The amount of human B7-H3 in the biological sample is inversely proportional to the amount of labeled rhB7-H3 standard bound to the anti-B7-H3 antibody. Similarly, human B7-H3 can also be assayed in biological fluids by a competition immunoassay utilizing rhB7-H3 standards labeled with a detectable substance and an unlabeled anti-human B7-H3 antibody.
In yet another aspect, this application provides a method for detecting the presence of B7-H3 in vivo (e.g., in vivo imaging in a subject). The subject method can be used to diagnose a disorder, e.g., a B7-H3-associated disorder. The method includes: (i) administering the anti-B7-H3 antibody or fragment thereof as described herein to a subject or a control subject under conditions that allow binding of the antibody or fragment to B7-H3; and (ii) detecting formation of a complex between the antibody or fragment and B7-H3, wherein a statistically significant change in the formation of the complex in the subject relative to the control subject is indicative of the presence of B7-H3. VI. Pharmaceutical Compositions
The invention also provides pharmaceutical compositions comprising an antibody, or antigen binding portion thereof, or ADC of the invention and a pharmaceutically acceptable carrier. The pharmaceutical compositions comprising antibodies or ADCs of the invention are for use in, but not limited to, diagnosing, detecting, or monitoring a disorder, in preventing, treating, managing, or ameliorating of a disorder or one or more symptoms thereof, and/or in research. In a specific embodiment, a composition comprises one or more antibodies of the invention. In another embodiment, the pharmaceutical composition comprises one or more antibodies or ADCs of the invention and one or more prophylactic or therapeutic agents other than antibodies or ADCs of the invention for treating a disorder in which B7-H3 activity is detrimental. Preferably, the prophylactic or therapeutic agents known to be useful for or having been or currently being used in the prevention, treatment, management, or amelioration of a disorder or one or more symptoms thereof. In accordance with these embodiments, the composition may further comprise of a carrier, diluent or excipient.
The antibodies and antibody-portions or ADCs of the invention can be incorporated into pharmaceutical compositions suitable for administration to a subject. Typically, the pharmaceutical composition comprises an antibody or antibody portion of the invention and a pharmaceutically acceptable carrier. As used herein,“pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Examples of pharmaceutically acceptable carriers include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like, as well as combinations thereof. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Pharmaceutically acceptable carriers may further comprise minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the antibody or antibody portion or ADC.
Various delivery systems are known and can be used to administer one or more antibodies or ADCs of the invention or the combination of one or more antibodies of the invention and a prophylactic agent or therapeutic agent useful for preventing, managing, treating, or ameliorating a disorder or one or more symptoms thereof, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the antibody or antibody fragment, receptor- mediated endocytosis (see, e.g., Wu and Wu, J. Biol. Chem.262:4429-4432 (1987)), construction of a nucleic acid as part of a retroviral or other vector, etc. Methods of administering a prophylactic or therapeutic agent of the invention include, but are not limited to, parenteral administration (e.g., intradermal, intramuscular, intraperitoneal, intravenous and subcutaneous), epidural administration, intratumoral administration, and mucosal administration (e.g., intranasal and oral routes). In addition, pulmonary administration can be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent. See, e.g., U.S. Pat. Nos.6,019,968, 5,985, 320, 5,985,309, 5,934, 272, 5,874,064, 5,855,913, 5,290, 540, and 4,880,078; and PCT Publication Nos. WO 92/19244, WO 97/32572, WO 97/44013, WO 98/31346, and WO 99/66903, each of which is incorporated herein by reference their entireties. In one embodiment, an antibody of the invention, combination therapy, or a composition of the invention is administered using Alkermes AIR® pulmonary drug delivery technology (Alkermes, Inc., Cambridge, Mass.). In a specific embodiment, prophylactic or therapeutic agents of the invention are administered intramuscularly, intravenously, intratumorally, orally, intranasally pulmonary or subcutaneously The prophylactic or therapeutic agents may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
In a specific embodiment, it may be desirable to administer the prophylactic or therapeutic agents of the invention locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion, by injection, or by means of an implant, said implant being of a porous or non-porous material, including membranes and matrices, such as sialastic membranes, polymers, fibrous matrices (e.g., Tissuel®), or collagen matrices. In one embodiment, an effective amount of one or more antibodies of the invention antagonists is administered locally to the affected area to a subject to prevent, treat, manage, and/or ameliorate a disorder or a symptom thereof. In another embodiment, an effective amount of one or more antibodies of the invention is
administered locally to the affected area in combination with an effective amount of one or more therapies (e.g., one or more prophylactic or therapeutic agents) other than an antibody of the invention of a subject to prevent, treat, manage, and/or ameliorate a disorder or one or more symptoms thereof.
In another embodiment, the prophylactic or therapeutic agent of the invention can be delivered in a controlled release or sustained release system. In one embodiment, a pump may be used to achieve controlled or sustained release (see Langer, supra; Sefton, 1987, CRC Crit. Ref. Biomed. Eng.14:20; Buchwald et al., 1980, Surgery 88:507; Saudek et al., 1989, N. Engl. J. Med.321:574). In another embodiment, polymeric materials can be used to achieve controlled or sustained release of the therapies of the invention (see e.g., Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla. (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, 1983, J., Macromol. Sci. Rev. Macromol. Chem.23:61; see also Levy et al., 1985, Science 228:190; During et al., 1989, Ann. Neurol.25:351; Howard et al., 1989, J. Neurosurg.71:105); U.S. Pat. No.5,679,377; U.S. Pat. No.5, 916,597; U. S. Pat. No.5,912,015; U.S. Pat. No.5,989,463; U.S. Pat. No.5,128,326; PCT Publication No. WO 99/15154; and PCT Publication No. WO 99/20253. Examples of polymers used in sustained release formulations include, but are not limited to, poly(2-hydroxy ethyl methacrylate), poly(methyl methacrylate), poly(acrylic acid), poly(ethylene-co-vinyl acetate), poly(methacrylic acid), polyglycolides (PLG), polyanhydrides, poly(N- vinyl pyrrolidone), poly(vinyl alcohol), polyacrylamide, poly(ethylene glycol), polylactides (PLA), poly(lactide-co-glycolides) (PLGA), and polyorthoesters. In a preferred embodiment, the polymer used in a sustained release formulation is inert, free of leachable impurities, stable on storage, sterile, and biodegradable. In yet another embodiment, a controlled or sustained release system can be placed in proximity of the prophylactic or therapeutic target, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol.2, pp.115-138 (1984)). Controlled release systems are discussed in the review by Langer (1990, Science 249:1527- 1533). Any technique known to one of skill in the art can be used to produce sustained release formulations comprising one or more therapeutic agents of the invention. See, e.g., U. S. Pat. No. 4,526, 938, PCT publication WO 91/05548, PCT publication WO 96/20698, Ning et al., 1996, “Intratumoral Radioimmunotherapy of a Human Colon Cancer Xenograft Using a Sustained-Release Gel,” Radiotherapy & Oncology 39:179-189, Song et al., 1995,“Antibody Mediated Lung Targeting of Long- Circulating Emulsions,” PDA Journal of Pharmaceutical Science & Technology 50:372-397, Cleek et al., 1997,“Biodegradable Polymeric Carriers for a bFGF Antibody for Cardiovascular Application,” Pro. Int'l. Symp. Control. Rel. Bioact. Mater.24:853-854, and Lam et al., 1997, “Microencapsulation of Recombinant Humanized Monoclonal Antibody for Local Delivery,” Proc. Int'l. Symp. Control Rel. Bioact. Mater.24:759- 760, each of which is incorporated herein by reference in their entireties.
In a specific embodiment, where the composition of the invention is a nucleic acid encoding a prophylactic or therapeutic agent, the nucleic acid can be administered in vivo to promote expression of its encoded prophylactic or therapeutic agent, by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (see U. S. Pat. No.4,980,286), or by direct injection, or by use of microparticle bombardment (e.g., a gene gun; Biolistic, Dupont), or coating with lipids or cell-surface receptors or transfecting agents, or by administering it in linkage to a homeobox-like peptide which is known to enter the nucleus (see, e.g., Joliot et al., 1991, Proc. Natl. Acad. Sci. USA 88:1864-1868). Alternatively, a nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression by homologous recombination.
A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include, but are not limited to, parenteral, e.g., intravenous, intradermal, subcutaneous, oral, intranasal (e.g., inhalation), transdermal (e.g., topical), transmucosal, and rectal administration. In a specific embodiment, the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous, subcutaneous, intramuscular, oral, intranasal, or topical administration to human beings. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection.
If the method of the invention comprises intranasal administration of a composition, the composition can be formulated in an aerosol form, spray, mist or in the form of drops. In particular, prophylactic or therapeutic agents for use according to the invention can be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant (e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas) In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges (composed of, e.g., gelatin) for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
If the method of the invention comprises oral administration, compositions can be formulated orally in the form of tablets, capsules, cachets, gel caps, solutions, suspensions, and the like. Tablets or capsules can be prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone, or hydroxypropyl methylcellulose); fillers (e.g., lactose, microcrystalline cellulose, or calcium hydrogen phosphate); lubricants (e.g., magnesium stearate, talc, or silica); disintegrants (e.g., potato starch or sodium starch glycolate) ; or wetting agents (e.g., sodium lauryl sulphate). The tablets may be coated by methods well-known in the art. Liquid preparations for oral administration may take the form of, but not limited to, solutions, syrups or suspensions, or they may be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, cellulose derivatives, or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters, ethyl alcohol, or fractionated vegetable oils); and preservatives (e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid). The preparations may also contain buffer salts, flavoring, coloring, and sweetening agents as appropriate. Preparations for oral administration may be suitably formulated for slow release, controlled release, or sustained release of a prophylactic or therapeutic agent(s).
The method of the invention may comprise pulmonary administration, e.g., by use of an inhaler or nebulizer, of a composition formulated with an aerosolizing agent. See, e.g., U.S. Pat. Nos. 6,019, 968, 5,985, 320, 5, 985,309, 5,934,272, 5,874,064, 5,855,913, 5,290,540, and 4,880,078; and PCT Publication Nos. WO 92/19244, WO 97/32572, WO 97/44013, WO 98/31346, and WO
99/66903, each of which is incorporated herein by reference their entireties. In a specific embodiment, an antibody of the invention, combination therapy, and/or composition of the invention is
administered using Alkermes AIR® pulmonary drug delivery technology (Alkermes, Inc., Cambridge, Mass.).
The method of the invention may comprise administration of a composition formulated for parenteral administration by injection (e.g., by bolus injection or continuous infusion). Formulations for injection may be presented in unit dosage form (e.g., in ampoules or in multi-dose containers) with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle (e.g., sterile pyrogen-free water) before use.
The methods of the invention may additionally comprise of administration of compositions formulated as depot preparations Such long acting formulations may be administered by implantation (e.g., subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compositions may be formulated with suitable polymeric or hydrophobic materials (e.g., as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives (e.g., as a sparingly soluble salt).
The methods of the invention encompass administration of compositions formulated as neutral or salt forms. Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2- ethylamino ethanol, histidine, procaine, etc.
Generally, the ingredients of compositions are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the mode of administration is infusion, composition can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the mode of administration is by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
In particular, the invention also provides that one or more of the prophylactic or therapeutic agents, or pharmaceutical compositions of the invention is packaged in a hermetically sealed container such as an ampoule or sachette indicating the quantity of the agent. In one embodiment, one or more of the prophylactic or therapeutic agents, or pharmaceutical compositions of the invention is supplied as a dry sterilized lyophilized powder or water free concentrate in a hermetically sealed container and can be reconstituted (e.g., with water or saline) to the appropriate concentration for administration to a subject. Preferably, one or more of the prophylactic or therapeutic agents or pharmaceutical compositions of the invention is supplied as a dry sterile lyophilized powder in a hermetically sealed container at a unit dosage of at least 5 mg, at least 10 mg, at least 15 mg, at least 25 mg, at least 35 mg, at least 45 mg, at least 50 mg, at least 75 mg, or at least 100 mg. The lyophilized prophylactic or therapeutic agents or pharmaceutical compositions of the invention should be stored at between 2° C. and 8° C. in its original container and the prophylactic or therapeutic agents, or pharmaceutical compositions of the invention should be administered within 1 week, within 5 days, within 72 hours, within 48 hours, within 24 hours, within 12 hours, within 6 hours, within 5 hours, within 3 hours, or within 1 hour after being reconstituted. In an alternative embodiment, one or more of the prophylactic or therapeutic agents or pharmaceutical compositions of the invention is supplied in liquid form in a hermetically sealed container indicating the quantity and concentration of the agent. Preferably, the liquid form of the administered composition is supplied in a hermetically sealed container at least 0.25 mg/ml, at least 0.5 mg/ml, at least 1 mg/ml, at least 2.5 mg/ml, at least 5 mg/ml, at least 8 mg/ml, at least 10 mg/ml, at least 15 mg/kg, at least 25 mg/ml, at least 50 mg/ml, at least 75 mg/ml or at least 100 mg/ml. The liquid form should be stored at between 2° C. and 8° C. in its original container.
The antibodies and antibody-portions of the invention can be incorporated into a
pharmaceutical composition suitable for parenteral administration. Preferably, the antibody or antibody-portions will be prepared as an injectable solution containing 0.1-250 mg/ml antibody. The injectable solution can be composed of either a liquid or lyophilized dosage form in a flint or amber vial, ampule or pre-filled syringe. The buffer can be L-histidine (1-50 mM), optimally 5-10 mM, at pH 5.0 to 7.0 (optimally pH 6.0). Other suitable buffers include but are not limited to, sodium succinate, sodium citrate, sodium phosphate or potassium phosphate. Sodium chloride can be used to modify the toxicity of the solution at a concentration of 0-300 mM (optimally 150 mM for a liquid dosage form). Cryoprotectants can be included for a lyophilized dosage form, principally 0-10% sucrose (optimally 0.5-1.0%). Other suitable cryoprotectants include trehalose and lactose. Bulking agents can be included for a lyophilized dosage form, principally 1-10% mannitol (optimally 2-4%). Stabilizers can be used in both liquid and lyophilized dosage forms, principally 1-50 mM L- methionine (optimally 5-10 mM). Other suitable bulking agents include glycine, arginine, can be included as 0-0.05% polysorbate-80 (optimally 0.005-0.01%). Additional surfactants include but are not limited to polysorbate 20 and BRIJ surfactants. The pharmaceutical composition comprising the antibodies and antibody-portions of the invention prepared as an injectable solution for parenteral administration, can further comprise an agent useful as an adjuvant, such as those used to increase the absorption, or dispersion of a therapeutic protein (e.g., antibody). A particularly useful adjuvant is hyaluronidase, such as Hylenex® (recombinant human hyaluronidase). Addition of hyaluronidase in the injectable solution improves human bioavailability following parenteral administration, particularly subcutaneous administration. It also allows for greater injection site volumes (i.e. greater than 1 ml) with less pain and discomfort, and minimum incidence of injection site reactions. (see WO2004078140, US2006104968 incorporated herein by reference).
The compositions of this invention may be in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes and suppositories. The preferred form depends on the intended mode of administration and therapeutic application. Typical preferred compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans with other antibodies. The preferred mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular). In a preferred embodiment, the antibody is administered by intravenous infusion or injection. In another preferred embodiment, the antibody is administered by intramuscular or subcutaneous injection.
Therapeutic compositions typically must be sterile and stable under the conditions of manufacture and storage. The composition can be formulated as a solution, microemulsion, dispersion, liposome or other ordered structure suitable to high drug concentration Sterile injectable solutions can be prepared by incorporating the active compound (i.e., antibody or antibody portion) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile, lyophilized powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and spray-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. The proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prolonged absorption of injectable compositions can be brought about by including, in the composition, an agent that delays absorption, for example, monostearate salts and gelatin.
The antibodies and antibody-portions or ADCs of the invention can be administered by a variety of methods known in the art, although for many therapeutic applications, the preferred route/mode of administration is subcutaneous injection, intravenous injection or infusion. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results. In certain embodiments, the active compound may be prepared with a carrier that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen,
polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J.R. Robinson, ed., Marcel Dekker, Inc., New York, 1978.
In certain embodiments, an antibody or antibody portion or ADC of the invention may be orally administered, for example, with an inert diluent or an assimilable edible carrier. The compound (and other ingredients, if desired) may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet. For oral therapeutic administration, the compounds may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. To administer a compound of the invention by other than parenteral administration, it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation.
In other embodiments, an antibody or antibody portion or ADC of the invention may be conjugated to a polymer-based species such that said polymer-based species may confer a sufficient size upon said antibody or antibody portion of the invention such that said antibody or antibody portion of the invention benefits from the enhanced permeability and retention effect (EPR effect) (See also PCT Publication No. WO2006/042146A2 and U.S. Publication Nos.2004/0028687A1, 2009/0285757A1, and 2011/0217363A1, and U.S. Patent No.7,695,719 (each of which is incorporated by reference herein in its entirety and for all purposes).
Supplementary active compounds can also be incorporated into the compositions. In certain embodiments, an antibody or antibody portion or ADC of the invention is formulated with and/or co- administered with one or more additional therapeutic agents that are useful for treating disorders in which B7-H3 activity is detrimental. For example, an anti-hB7-H3 antibody or antibody portion or ADC of the invention may be formulated and/or co-administered with one or more additional antibodies that bind other targets (e.g., antibodies that bind cytokines or that bind cell surface molecules). Furthermore, one or more antibodies of the invention may be used in combination with two or more of the foregoing therapeutic agents. Such combination therapies may advantageously utilize lower dosages of the administered therapeutic agents, thus avoiding possible toxicities or complications associated with the various monotherapies.
In certain embodiments, an antibody or ADC to B7-H3 or fragment thereof is linked to a half-life extending vehicle known in the art. Such vehicles include, but are not limited to, the Fc domain, polyethylene glycol, and dextran. Such vehicles are described, e.g., in U.S. Application Serial No.09/428,082 and published PCT Application No. WO 99/25044, which are hereby incorporated by reference for any purpose.
It will be readily apparent to those skilled in the art that other suitable modifications and adaptations of the methods of the invention described herein are obvious and may be made using suitable equivalents without departing from the scope of the invention or the embodiments disclosed herein. Having now described the invention in detail, the same will be more clearly understood by reference to the following examples, which are included for purposes of illustration only and are not intended to be limiting EXAMPLES
Example 1: Synthesis of Exemplary Bcl-xL Inhibitors
This Example provides synthetic methods for exemplary Bcl-xL inhibitory compounds W3.01-W3.43. Bcl-xL inhibitors (W3.01-W3.43) and synthons (Examples 2.1-2.72) were named using ACD/Name 2012 release (Build 56084, 05 April 2012, Advanced Chemistry Development Inc., Toronto, Ontario), ACD/Name 2014 release (Build 66687, 25 October 2013, Advanced Chemistry Development Inc., Toronto, Ontario), ChemDraw® Ver.9.0.7 (CambridgeSoft, Cambridge, MA), ChemDraw® Ultra Ver.12.0 (CambridgeSoft, Cambridge, MA), or ChemDraw® Professional Ver. 15.0.0.106. Bcl-xL inhibitor and synthon intermediates were named with ACD/Name 2012 release (Build 56084, 05 April 2012, Advanced Chemistry Development Inc., Toronto, Ontario), ACD/Name 2014 release (Build 66687, 25 October 2013, Advanced Chemistry Development Inc., Toronto, Ontario), ChemDraw® Ver.9.0.7 (CambridgeSoft, Cambridge, MA), ChemDraw® Ultra Ver.12.0 (CambridgeSoft Cambridge MA) or ChemDraw® Professional Ver 1500106 1.1. Synthesis of 6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4- tetrahydroquinolin-7-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H- pyrazol-4-yl]pyridine-2-carboxylic acid (Compound W3.01)
1.1.1. 3-bromo-5,7-dimethyladamantanecarboxylic acid
To a 50 mL round-bottomed flask at 0 °C was added bromine (16 mL). Iron powder (7 g) was added, and the reaction was stirred at 0 °C for 30 minutes. 3,5-Dimethyladamantane-1- carboxylic acid (12 g) was then added. The mixture was then warmed to room temperature and stirred for 3 days. An ice/concentrated HCl mixture was poured into the reaction mixture. The resulting suspension was treated twice with Na2SO3 (50 g in 200 mL water) and extracted three times with dichloromethane. The combined organic layers were washed with 1N aqueous HCl, dried over Na2SO4, filtered, and concentrated to give the crude title compound.
1.1.2. 3-bromo-5,7-dimethyladamantanemethanol
To a solution of Example 1.1.1 (15.4 g) in tetrahydrofuran (200 mL) was added BH3 (1M in tetrahydrofuran, 150 mL). The mixture was stirred at room temperature overnight. The reaction mixture was then carefully quenched via dropwise addition of methanol. The mixture was then concentrated under vacuum and the residue was partitioned between ethyl acetate (500 mL) and 2N aqueous HCl (100 mL). The aqueous layer was further extracted twice with ethyl acetate and the combined organic extracts were combined and washed with water and brine, and dried over Na2SO4. Filtration and evaporation of the solvent gave the title compound.
1.1.3. 1-((3-bromo-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl)-1H- pyrazole
To a solution of Example 1.1.2 (8.0 g) in toluene (60 mL) was added 1H-pyrazole (1.55 g) and cyanomethylenetributylphosphorane (2.0 g). The mixture was stirred at 90 oC overnight. The reaction mixture was then concentrated and the residue was purified by silica gel column
chromatography (10:1 hexane:ethyl acetate) to provide the title compound. MS (ESI) m/e 324.2 (M+H)+.
1.1.4. 2-{[3,5-dimethyl-7-(1H-pyrazol-1-ylmethyl)tricyclo[3.3.1.13,7]dec- 1-yl]oxy}ethanol
To a solution of Example 1.1.3 (4.0 g) in ethane-1,2-diol (12 mL) was added triethylamine (3 mL). The mixture was stirred at 150 oC under microwave conditions (Biotage) for 45 minutes. The mixture was poured into water (100 mL) and extracted three times with ethyl acetate. The combined organic extracts were washed with water and brine, and dried over Na2SO4. Filtration and evaporation of the solvent gave the crude title compound which was purified via column chromatography, eluting with 20% ethyl acetate in hexane followed by 5% methanol in dichloromethane, to provide the title compound. MS (ESI) m/e 305.2 (M+H)+. 1.1.5. 2-({3,5-dimethyl-7-[(5-methyl-1H-pyrazol-1- yl)methyl]tricyclo[3.3.1.13,7]dec-1-yl}oxy)ethanol
To a cooled (-78 oC) solution of Example 1.1.4 (6.05 g) in tetrahydrofuran (100 mL) was added n-BuLi (40 mL, 2.5M in hexane). The mixture was stirred at -78 oC for 1.5 hours. Then, iodomethane (10 mL) was added through a syringe and the mixture was stirred at -78 oC for 3 hours. The reaction mixture was then quenched with aqueous NH4Cl and extracted twice with ethyl acetate, and the combined organic extracts were washed with water and brine. After drying over Na2SO4, the solution was filtered and concentrated and the residue was purified by silica gel column
chromatography (5% methanol in dichloromethane) to provide the title compound. MS (ESI) m/e 319.5 (M+H)+.
1.1.6. 1-({3,5-dimethyl-7-[2-(hydroxy)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-4-iodo-5-methyl-1H-pyrazole
To a solution of Example 1.1.5 (3.5 g) in N,N-dimethylformamide (30 mL) was added N- iodosuccinimide (3.2 g). The mixture was stirred at room temperature for 1.5 hours. The reaction mixture was then diluted with ethyl acetate (600 mL) and washed with aqueous NaHSO3, water, and brine. After drying over Na2SO4, the solution was filtered and concentrated and the residue was purified by silica gel chromatography (20% ethyl acetate in dichloromethane) to give the title compound. MS (ESI) m/e 445.3 (M+H)+.
1.1.7. 2-((3-((4-iodo-5-methyl-1H-pyrazol-1-yl)methyl)-5,7- dimethyladamantan-1-yl)oxy)ethyl methanesulfonate
To a cooled solution (0 °C) of Example 1.1.6 (5.45 g) in dichloromethane (100 mL) was added triethylamine (5.13 mL) and methanesulfonyl chloride (0.956 mL). The mixture was stirred at room temperature for 1.5 hours, diluted with ethyl acetate (600 mL) and washed with water (120 mL) and brine (120 mL). The organic layer was dried over Na2SO4, filtered, and concentrated to provide the title compound. MS (ESI) m/e 523.4 (M+H)+.
1.1.8. 2-((3-((4-iodo-5-methyl-1H-pyrazol-1-yl)methyl)-5,7- dimethyladamantan-1-yl)oxy)-N-methylethanamine
A solution of Example 1.1.7 (6.41 g) in 2M methylamine in ethanol (15 mL) was stirred at overnight and concentrated. The residue was diluted with ethyl acetate and washed with aqueous NaHCO3, water and brine. The organic layer was dried over Na2SO4, filtered, and concentrated to provide the title compound. MS (ESI) m/e 458.4 (M+H)+.
1.1.9. tert-butyl [2-({3-[(4-iodo-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl]methylcarbamate
To a solution of Example 1.1.8 (2.2 g) in tetrahydrofuran (30 mL) was added di-tert-butyl dicarbonate (1.26 g) and a catalytic amount of 4-dimethylaminopyridine. The mixture was stirred at room temperature for 1.5 hours and then diluted with ethyl acetate (300 mL). The solution was washed with saturated aqueous NaHCO3 water (60 mL) and brine (60 mL) The organic layer was dried with Na2SO4, filtered and concentrated. The residue was purified by silica gel chromatography, eluting with 20% ethyl acetate in dichloromethane, to provide the title compound. MS (ESI) m/e 558.5 (M+H)+.
1.1.10. tert-butyl (2-((3,5-dimethyl-7-((5-methyl-4-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)-1H-pyrazol-1-yl)methyl)adamantan-1- yl)oxy)ethyl)(methyl)carbamate
To a solution of Example 1.1.9 (1.2 g) in dioxane was added bis(benzonitrile)palladium(II) chloride (0.04 g), 4,4,5,5-tetramethyl-1,3,2-dioxaborolane (0.937 mL) and triethylamine (0.9 mL). The mixture was heated at reflux overnight, diluted with ethyl acetate and washed with water (60 mL) and brine (60 mL). The organic layer was dried over Na2SO4, filtered and concentrated to provide the title compound. MS (ESI) m/e 558.5 (M+H)+.
1.1.11. tert-butyl 3-(1-((3-(2-((tert-butoxycarbonyl)(methyl)amino) ethoxy)-5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H- pyrazol-4-yl)-6-chloropicolinate
To Example 1.1.10 (100 mg) and tert-butyl 3-bromo-6-chloropicolinate (52.5 mg) in dioxane (2 mL) was added tris(dibenzylideneacetone)dipalladium(0) (8.2 mg), K3PO4 (114 mg), 1,3,5,7- tetramethyl-8-phenyl-2,4,6-trioxa-8-phosphaadamantane (5.24 mg) and water (0.8 mL). The mixture was stirred at 95 °C for 4 hours, diluted with ethyl acetate and washed with water and brine. The organic layer was dried over Na2SO4, filtered, concentrated and purified by flash chromatography, eluting with 20% ethyl acetate in heptanes and then with 5% methanol in dichloromethane, to provide the title compound. MS (ESI) m/e 643.3 (M+H)+.
1.1.12. tert-butyl 3-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan- 1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(1,2,3,4- tetrahydroquinolin-7-yl)picolinate
A mixture of Example 1.1.11 (480 mg), 7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 1,2,3,4-tetrahydroquinoline (387 mg), dichlorobis(triphenylphosphine)-palladium(II) (78 mg) and CsF (340 mg) in dioxane (12 mL) and water (5 mL) was heated at 100 oC for 5 hours. After this time the reaction mixture was allowed to cool to room temperature and then diluted with ethyl acetate. The resulting mixture was washed with water and brine, and the organic layer was dried over Na2SO4, filtered, and concentrated. The residue was purified by flash chromatography, eluting with 50% ethyl acetate in heptanes to provide the title compound. MS (APCI) m/e 740.4 (M+H)+.
1.1.13. tert-butyl 6-(1-(benzo[d]thiazol-2-ylcarbamoyl)-1,2,3,4- tetrahydroquinolin-7-yl)-3-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan- 1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate To a solution of benzo[d]thiazol-2-amine (114 mg) in acetonitrile (5 mL) was added bis(2,5- dioxopyrrolidin-1-yl) carbonate (194 mg). The mixture was stirred for 1 hour, and Example 1.1.12 (432 mg) in acetonitrile (5 mL) was added. The mixture was stirred overnight, diluted with ethyl acetate, washed with water and brine, and the organic layer was dried over Na2SO4, filtered, and concentrated. The residue was purified by flash chromatography, eluting with 50% ethyl acetate in heptanes to provide the title compound.
1.1.14. 6-(1-(benzo[d]thiazol-2-ylcarbamoyl)-1,2,3,4-tetrahydroquinolin- 7-yl)-3-(1-((3,5-dimethyl-7-(2-(methylamino)ethoxy)adamantan- 1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinic acid
Example 1.1.13 (200 mg) in dichloromethane (5 mL) was treated with trifluoroacetic acid (2.5 mL) overnight. The mixture was concentrated to provide the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 8.40 (s, 1H), 8.30 (s, 2H), 8.02 (d, 1H), 7.85 (d, 1H), 7.74-7.83 (m, 2H), 7.42-7.53 (m, 2H), 7.38 (t, 1H), 7.30 (d, 1H), 7.23 (t, 1H), 3.93-4.05 (m, 2H), 3.52-3.62 (m, 2H), 2.97-3.10 (m, 2H), 2.84 (t, 2H), 2.56 (t, 2H), 2.23 (s, 3H), 1.88-2.00 (m, 2H), 1.45 (s, 2H), 1.25-1.39 (m, 4H), 1.12-1.22 (m, 4H), 1.00-1.09 (m, 2H), 0.89 (s, 6H). MS (ESI) m/e 760.1 (M+H)+.
1.2. Synthesis of 6-[4-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-2H-1,4- benzoxazin-6-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H- pyrazol-4-yl]pyridine-2-carboxylic acid (Compound W3.02)
1.2.1. tert-butyl 3-(1-(((--3-(2-((tert-butoxycarbonyl)(methyl)
amino)ethoxy)-5,7-dimethyladamantan-1-yl)methyl)-5-methyl- 1H-pyrazol-4-yl)-6-(3,4-dihydro-2H-benzo[b][1,4]oxazin-6- yl)picolinate
To a solution of 6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,4-dihydro-2H- benzo[b][1,4]oxazine (122 mg) in dioxane (4 mL) and water (1 mL) was added Example 1.1.11 ( 300 mg), bis(triphenylphosphine)palladium(II) dichloride (32.7 mg), and CsF (212 mg). The mixture was stirred at reflux overnight. The mixture was diluted with ethyl acetate (500 mL) and washed with water, brine and dried over Na2SO4. Filtration and evaporation of the solvents gave crude material which was purified via column chromatography (20% ethyl acetate in heptane followed by 5% methanol in dichloromethane) to provide the title compound. MS (ESI) m/e 742.4 (M+H)+.
1.2.2. 6-[4-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-2H-1,4- benzoxazin-6-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5- methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid
To an ambient suspension of bis(2,5-dioxopyrrolidin-1-yl) carbonate (70.4 mg) in acetonitrile (4 mL) was added benzo[d]thiazol-2-amine (41.3 mg) and the mixture was stirred for one hour. A solution of Example 121 (170 mg) in acetonitrile (1 mL) and water (10 mL) was added and the suspension was stirred vigorously overnight. The mixture was diluted with ethyl acetate (500 mL) and washed with water, brine and dried over Na2SO4. Filtration and evaporation of the solvents afforded a residue which was loaded on a column and eluted with 20% ethyl acetate in heptane followed by 5% methanol in dichloromethane. The resultant material was treated with 20% TFA in dichloromethane overnight. After evaporation of the solvent, the residue was purified via HPLC (Gilson system, eluting with 10- 85% acetonitrile in 0.1% TFA in water) to provide the title compound.1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 8.76 (s, 1H), 8.24-8.46 (m, 2H), 7.97 (d, 1H), 7.70-7.89 (m, 3H), 7.47 (s, 1H), 7.35-7.47 (m, 2H), 7.24 (t, 1H), 7.02 (d, 1H), 4.32-4.42 (m, 3H), 4.14-4.23 (m, 3H), 3.90 (s, 3H), 3.57 (t, 3H), 2.93-3.11 (m, 2H), 2.57 (t, 3H), 2.23 (s, 3H), 1.46 (s, 2H), 1.24-1.39 (m, 4H), 0.98-1.25 (m, 5H), 0.89 (s, 6H). MS (ESI) m/e 760.4 (M+H)+.
1.3. Synthesis of 6-[4-(1,3-benzothiazol-2-ylcarbamoyl)-1-methyl-1,2,3,4- tetrahydroquinoxalin-6-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H- pyrazol-4-yl]pyridine-2-carboxylic acid (Compound W3.03)
1.3.1. tert-butyl 3-(1-((3-(2-((tert-butoxycarbonyl)(methyl)amino) ethoxy)-5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H- pyrazol-4-yl)-6-(1-methyl-1,2,3,4-tetrahydroquinoxalin-6- yl)picolinate
To a solution of 1-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,2,3,4- tetrahydroquinoxaline (140 mg) in dioxane (4 mL) and water (1 mL) was added Example 1.1.11 ( 328 mg), bis(triphenylphosphine)palladium(II) dichloride (35.8 mg), and CsF (232 mg). The mixture was stirred at reflux overnight. The mixture was diluted with ethyl acetate (500 mL) and washed with water, brine and dried over Na2SO4. Filtration and evaporation of the solvent gave crude material which was purified via column chromatography, eluting with 20% ethyl acetate in heptane followed by 5% methanol in dichloromethane, to provide the title compound. MS (ESI) m/e 755.5 (M+H)+.
1.3.2. 6-[4-(1,3-benzothiazol-2-ylcarbamoyl)-1-methyl-1,2,3,4- tetrahydroquinoxalin-6-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5- methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid
To an ambient suspension of bis(2,5-dioxopyrrolidin-1-yl) carbonate (307 mg) in acetonitrile (10 mL) was added benzo[d]thiazol-2-amine (180 mg) and the mixture was stirred for one hour. A solution of Example 1.3.1 (600 mg) in acetonitrile (3 mL) was added, and the suspension was vigorously stirred overnight. The mixture was diluted with ethyl acetate (500 mL) and washed with water and brine and dried over Na2SO4. Filtration and evaporation of the solvents afforded a residue which was loaded on a column and eluted with 20% ethyl acetate in heptane (1 L) followed by 5% methanol in dichloromethane. The resultant material was treated with 20% TFA in dichloromethane overnight After evaporation of solvent the residue was purified on an HPLC (Gilson system eluting with 10-85% acetonitrile in 0.1% TFA in water) to give the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 8.17-8.44 (m, 3H), 7.90 (d, 1H), 7.68-7.84 (m, 3H), 7.45 (s, 2H), 7.37 (t, 1H), 7.22 (t, 1H), 6.83 (d, 1H), 3.96-4.12 (m, 2H), 3.89 (s, 3H), 3.57 (t, 2H), 3.44 (t, 2H), 2.93-3.09 (m, 4H), 2.56 (t, 3H), 2.21 (s, 3H), 1.45 (s, 2H), 1.25-1.39 (m, 4H), 0.99-1.22 (m, 7H), 0.89 (s, 6 H). MS (ESI) m/e 760.4 (M+H)+.
1.4. Synthesis of 3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec- 1-yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[1-(1,3-benzothiazol-2- ylcarbamoyl)-5,6-dihydroimidazo[1,5-a]pyrazin-7(8H)-yl]pyridine-2- carboxylic acid (Compound W3.04)
1.4.1. 2-((3-((4-iodo-5-methyl-1H-pyrazol-1-yl)methyl)-5,7- dimethyladamantan-1-yl)oxy)ethanamine
A solution of Example 1.1.7 (4.5 g) in 7N ammonium in methanol (15 mL) was stirred at 100 oC for 20 minutes under microwave conditions (Biotage Initiator). The reaction mixture was concentrated under vacuum. The residue was diluted with ethyl acetate (400 mL) and washed with aqueous NaHCO3, water (60 mL) and brine (60 mL). The organic layer was dried (anhydrous Na2SO4), the solution was filtered and concentrated, and the residue was used in the next reaction without further purification. MS (ESI) m/e 444.2 (M+H)+.
1.4.2. tert-butyl (2-((3-((4-iodo-5-methyl-1H-pyrazol-1-yl)methyl)-5,7- dimethyladamantan-1-yl)oxy)ethyl)carbamate
To a solution of Example 1.4.1 (4.4 g) in tetrahydrofuran (100 mL) was added di-tert-butyl dicarbonate (2.6 g) and N,N-dimethyl-4-aminopyridine (100 mg). The mixture was stirred for 1.5 hours. The reaction mixture was diluted with ethyl acetate (300 mL) and washed with aqueous NaHCO3, water (60 mL) and brine (60 mL). After drying (anhydrous Na2SO4), the solution was filtered and concentrated, and the residue was purified by silica gel column chromatography (20% ethyl acetate in dichloromethane) to give the title compound. MS (ESI) m/e 544.2 (M+H)+.
1.4.3. 6-fluoro-3-bromopicolinic acid
A slurry of 6-amino-3-bromopicolinic acid (25 g) in 400 mL 1:1 dichloromethane/chloroform was added to nitrosonium tetrafluoroborate (18.2 g) in dichloromethane (100 mL) at 5 oC over 1 hour. The resulting mixture was stirred for another 30 minutes, warmed to 35 oC, and stirred overnight. The reaction mixture was cooled to room temperature and adjusted to pH 4 with a NaH2PO4 solution. The resulting solution was extracted three times with dichloromethane, and the combined extracts were washed with brine, dried over sodium sulfate, filtered and concentrated to provide the title compound.
1.4.4. Tert-butyl 3-bromo-6-fluoropicolinate
Para-toluenesulfonyl chloride (27.6 g) was added to a solution of Example 1.4.3 (14.5 g), pyridine (26.7 mL) and tert-butanol (80 mL) in dichloromethane (100 mL) at 0 °C. The reaction was stirred for 15 minutes, warmed to room temperature, and stirred overnight. The solution was concentrated and partitioned between ethyl acetate and Na2CO3 solution The layers were separated and the aqueous layer was extracted with ethyl acetate. The organic layers were combined, rinsed with Na2CO3 solution and brine, dried over sodium sulfate, filtered, and concentrated to provide the title compound.
1.4.5. Ethyl 7-(5-bromo-6-(tert-butoxycarbonyl)pyridin-2-yl)-5,6,7,8- tetrahydroimidazo[1,5-a]pyrazine-1-carboxylate
Ethyl 5,6,7,8-tetrahydroimidazo[1,5-a]pyrazine-1-carboxylate hydrochloride (692 mg) and Example 1.4.4 (750 mg) were dissolved in dimethyl sulfoxide (6 mL). N,N-Diisopropylethylamine (1.2 mL) was added, and the solution was heated at 50 °C for 16 hours. The solution was cooled, diluted with water (20 mL), and extracted with ethyl acetate (50 mL). The organic portion was washed with brine and dried on anhydrous sodium sulfate. The solution was concentrated and, upon standing for 16 hours, solid crystals formed. The crystals were washed with diethyl ether to yield the title compound. MS (ESI) m/e 451, 453 (M+H)+, 395, 397 (M-tert-butyl)+.
1.4.6. Ethyl 7-(6-(tert-butoxycarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyridin-2-yl)-5,6,7,8-tetrahydroimidazo[1,5- a]pyrazine-1-carboxylate
The title compound was prepared by substituting Example 1.4.5 for Example 1.1.9 in Example 1.1.10. MS (ESI) m/e 499 (M+H)+, 443 (M- tert-butyl)+, 529 (M+MeOH-H)-.
1.4.7. Ethyl 7-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-5,6,7,8- tetrahydroimidazo[1,5-a]pyrazine-1-carboxylate
Example 1.4.6 (136 mg) and Example 1.4.2 (148 mg) were dissolved in 1,4-dioxane (3 mL) and water (0.85 mL). Tripotassium phosphate (290 mg) was added, and the solution was degassed and flushed with nitrogen three times. Tris(dibenzylideneacetone)dipalladium(0) (13 mg) and 1,3,5,7- tetramethyl-8-tetradecyl-2,4,6-trioxa-8-phosphaadamantane (12 mg) were added. The solution was degassed, flushed with nitrogen once, and heated to 70 °C for 16 hours. The reaction was cooled and diluted with ethyl acetate (10 mL) and water (3 mL). The layers were separated, and the organic layer was washed with brine and dried on anhydrous sodium sulfate. After filtration, the filtrate was concentrated and purified by flash column chromatography on silica gel, eluting with 5% methanol in ethyl acetate. The solvent was removed under reduced pressure to give the title compound. MS (ESI) m/e 760 (M+H)+, 758 (M-H)-.
1.4.8. 7-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-5,6,7,8- tetrahydroimidazo[1,5-a]pyrazine-1-carboxylic acid
Example 1.4.7 (200 mg) was dissolved in tetrahydrofuran (0.7 mL), methanol (0.35 mL), and water (035 mL) Lithium hydroxide monohydrate (21 mg) was added and the solution was stirred at room temperature for 16 hours. HCl (1M, 0.48 mL) was added and the water was removed by azeotroping twice with ethyl acetate (20 mL). The solvent was removed under reduced pressure, and the material was dried under vacuum. The material was dissolved in dichloromethane (5 mL) and ethyl acetate (1 mL) and dried over anhydrous sodium sulfate. After filtration, the solvent was removed under reduced pressure to give the title compound. MS (ESI) m/e 760 (M+H)+, 758 (M-H)-.
1.4.9. Tert-butyl 6-(1-(benzo[d]thiazol-2-ylcarbamoyl)-5,6- dihydroimidazo[1,5-a]pyrazin-7(8H)-yl)-3-(1-((3-(2-((tert- butoxycarbonyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate
Example 1.4.8 (160 mg) and benzo[d]thiazol-2-amine (35 mg) were dissolved in
dichloromethane (1.5 mL). 1-Ethyl-3-[3-(dimethylamino)propyl]-carbodiimide hydrochloride (85 mg) and 4-(dimethylamino)pyridine (54 mg) were added, and the solution was stirred at room temperature for 16 hours. The material was purified by flash column chromatography on silica gel, eluting with 2.5-5% methanol in ethyl acetate. The solvent was removed under reduced pressure to give the title compound. MS (ESI) m/e 892 (M+H)+, 890 (M-H)-.
1.4.10. 3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[1-(1,3-benzothiazol-2- ylcarbamoyl)-5,6-dihydroimidazo[1,5-a]pyrazin-7(8H)- yl]pyridine-2-carboxylic acid
The title compound was prepared by substituting Example 1.4.9 for Example 1.1.13 in Example 1.1.14. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 11.50 (bs, 1H), 8.21 (d, 1H), 7.98 (d, 1H), 7.93 (s, 1H), 7.76 (d, 1H), 7.66 (bs, 3H), 7.58 (d, 1H), 7.44 (t, 1H), 7.33 (s, 1H), 7.31 (t, 1H), 7.15 (d, 1H), 6.97 (d, 1H), 5.10 (s, 2H), 4.26 (m, 2H), 4.08 (t, 2H), 3.84 (s, 2H), 2.90 (m, 4H), 2.13 (s, 3H), 1.42 (s, 2H), 1.30 (q, 4H), 1.15 (m, 2H), 1.04 (q, 4H), 0.87 (s, 6H). MS (ESI) m/e 736 (M+H)+, 734 (M-H)-.
1.5. Synthesis of 3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec- 1-yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-5-hydroxy-3,4-dihydroisoquinolin-2(1H)-yl]pyridine-2- carboxylic acid (Compound W3.05)
1.5.1. tert-butyldiphenyl(vinyl)silane
The title compound was prepared as described in J Org Chem, 70(4), 1467 (2005).
1.5.2. 2-(tert-butyldiphenylsilyl)ethanol
Example 1.5.1 (8.2 g) was dissolved in tetrahydrofuran (30 mL), then a 0.5M solution of 9- borabicyclo[3.3.1]nonane in tetrahydrofuran (63 mL) was added and the reaction was stirred at room temperature for 2.5 hours. The reaction was warmed to 37 °C, then 3.0N aqueous NaOH (11 mL) was added, followed by the very careful dropwise addition of 30% aqueous H2O2 (11 mL). Once the peroxide addition was completed the reaction was stirred for one hour and water (200 mL) and diethyl ether (200 mL) were added. The organic layer was washed with brine and dried over sodium sulfate. After filtration and concentration, purification by silica gel chromatography, eluting with heptanes/ethyl acetate (3/1), gave the title compound.
1.5.3. 5-(2-(tert-butyldiphenylsilyl)ethoxy)isoquinoline
Triphenylphosphine (262 mg) was dissolved in tetrahydrofuran (2 mL). Example 1.5.2 (285 mg), isoquinolin-5-ol (121 mg), and diisopropyl azodicarboxylate (203 mg) were added. The reaction was stirred at room temperature for 30 minutes, then more isoquinolin-5-ol (41 mg) was added and the reaction was stirred overnight. The reaction was then concentrated and purification by flash chromatography, eluting with heptanes/ethyl acetate (83/17), gave the title compound. MS (DCI) m/e 412.2 (M+H)+.
1.5.4. 8-bromo-5-(2-(tert-butyldiphenylsilyl)ethoxy)isoquinoline Example 1.5.3 (6.2 g) was dissolved in acetic acid (40 mL), and sodium acetate (2.2 g) was added. A solution of bromine (0.70 mL) in acetic acid (13 mL) was added slowly. The reaction was stirred at room temperature overnight. The reaction was carefully added to 2M aqueous Na2CO3 and extracted with ethyl acetate. The organic layer was washed with brine and dried over sodium sulfate. After filtration and concentration, purification by silica gel chromatography, eluting with
heptanes/ethyl acetate (9/1), gave the title compound. MS (DCI) m/e 490.1, 492.1 (M+H)+.
1.5.5. 8-bromo-5-(2-(tert-butyldiphenylsilyl)ethoxy)-1,2,3,4- tetrahydroisoquinoline
Example 1.5.4 (4.46 g) was dissolved in methanol (45 mL). Sodium cyanoborohydride (2.0 g) was added followed by trifluoroborane etherate (4.0 mL, 31.6 mmol). The mixture was heated under reflux for two hours and then cooled to room temperature. Additional sodium cyanoborohydride (2.0 g) and trifluoroborane etherate (4.0 mL) were added, and the mixture was heated under reflux for two more hours. The reaction was cooled, then added to 1/1 water/2M aqueous Na2CO3 (150 mL). The mixture was extracted with dichloromethane (twice with 100 mL). The organic layer was dried over sodium sulfate. Filtration and concentration provided the title compound that was used in the next step with no further purification. MS (DCI) m/e 494.1, 496.1 (M+H)+.
1.5.6. tert-butyl 8-bromo-5-(2-(tert-butyldiphenylsilyl)ethoxy)-3,4- dihydroisoquinoline-2(1H)-carboxylate
Example 1.5.5 (3.9 g) was dissolved in dichloromethane (25 mL), and triethylamine (3.3 mL) and di-tert-butyl dicarbonate (1.9 g) were added. The reaction mixture was stirred at room temperature for three hours. The reaction was then concentrated and purified by flash
chromatography, eluting with heptanes/ethyl acetate (96/4), to provide the title compound.
1.5.7. 2-tert-butyl 8-methyl 5-(2-(tert-butyldiphenylsilyl)ethoxy)-3,4- dihydroisoquinoline-2,8(1H)-dicarboxylate
Example 1.5.6 (3.6 g) and [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II) dichloromethane (0025 g) were placed in a 250 mL SS pressure bottle and methanol (10 mL) and triethylamine (0.469 mL) were added. After degassing the reactor with argon several times, the flask was charged with carbon monoxide and heated to 100 °C for 16 hours at 40 psi. The reaction mixture was cooled, concentrated, and purified by flash silica gel chromatography, eluting heptanes/ethyl acetate (88/12), to provide the title compound.
1.5.8. methyl 5-(2-(tert-butyldiphenylsilyl)ethoxy)-1,2,3,4- tetrahydroisoquinoline-8-carboxylate
Example 1.5.7 (1.8 g) was dissolved in 4N HCl in dioxane (25 mL) and stirred at room temperature for 45 minutes. The reaction was then concentrated to provide the title compound as a hydrochloride salt. MS (DCI) m/e 474.2 (M+H)+.
1.5.9. methyl 2-(5-bromo-6-(tert-butoxycarbonyl)pyridin-2-yl)-5-(2- (tert-butyldiphenylsilyl)ethoxy)-1,2,3,4-tetrahydroisoquinoline-8- carboxylate
To a solution of Example 1.5.8 (1.6 g) and Example 1.4.4 (1.0 g) in dimethyl sulfoxide (6 mL) was added N,N-diisopropylethylamine (1.4 mL). The mixture was stirred at 50 oC for 24 hours. The mixture was then diluted with diethyl ether and washed with water and brine, and dried over Na2SO4. Filtration and evaporation of the solvent and silica gel column purification (eluting with 5% ethyl acetate in hexane) gave the title compound.
1.5.10. 1-((3-(2-azidoethoxy)-5,7-dimethyladamantan-1-yl)methyl)-4- iodo-5-methyl-1H-pyrazole
Example 1.1.6 (2 g) was dissolved in dichloromethane (20 mL), and triethylamine (0.84 mL) was added. After cooling the reaction solution to 5 °C, mesyl chloride (0.46 mL) was added dropwise. The cooling bath was removed and the reaction was stirred at room temperature for two hours. Saturated NaHCO3 was added, the layers were separated, and the organic layer was washed with brine, and dried over Na2SO4. After filtration and concentration, the residue was dissolved in N,N dimethylformamide (15 mL) and sodium azide (0.88 g) was added, and the reaction was heated to 80 °C for two hours. The reaction was then cooled to room temperature and poured into diethyl ether and water. The organic layer was separated and washed with brine and dried over Na2SO4. After filtration and concentration, purification by silica gel chromatography, eluting with
heptanes/ethyl acetate (4/1), gave the title compound. MS (DCI) m/e 470.0 (M+H)+.
1.5.11. methyl 2-(6-(tert-butoxycarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyridin-2-yl)-5-(2-(tert- butyldiphenylsilyl)ethoxy)-1,2,3,4-tetrahydroisoquinoline-8- carboxylate
Example 1.5.9 (1.5 g), 4,4,5,5-tetramethyl-1,3,2-dioxaborolane (0.46 mL), [1,1'- bis(diphenylphosphino)ferrocene]dichloropalladium(II) dichloromethane (86 mg), and triethylamine (0.59 mL) were dissolved in acetonitrile (6.5 mL) under a nitrogen atmosphere, then the reaction was heated under reflux overnight. The reaction was then cooled to room temperature and ethyl acetate and water were added. The organic layer was washed with brine and dried over Na2SO4. After filtration and concentration, purification by silica gel chromatography, using a gradient of 10-20% ethyl acetate in heptanes, gave the title compound. MS (ESI) m/e 777.1 (M+H)+.
1.5.12. methyl 2-(5-(1-((3-(2-azidoethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(tert- butoxycarbonyl)pyridin-2-yl)-5-(2-(tert-butyldiphenylsilyl)
ethoxy)-1,2,3,4-tetrahydroisoquinoline-8-carboxylate
Example 1.5.11 (1.22 g) and Example 1.5.10 (0.74 g) were dissolved in tetrahydrofuran (16 mL) under a nitrogen atmosphere, and tripotassium phosphate (4.5 g) and water (5 mL) were added. Tris(dibenzylideneacetone)dipalladium(0) (70 mg) and 1,3,5,7-tetramethyl-8-tetradecyl-2,4,6-trioxa- 8-phosphaadamantane (66 mg) were then added, the reaction was heated at reflux overnight, and then allowed to cool to room temperature. Ethyl acetate and water were then added, and the organic layer washed with brine and dried over Na2SO4. After filtration and concentration, the crude material was purified by silica gel chromatography, eluting with heptanes/ethyl acetate (7/3), gave the title compound. MS (DCI) m/e 992.3 (M+H)+.
1.5.13. 2-(5-(1-((3-(2-azidoethoxy)-5,7-dimethyladamantan-1-yl)methyl)- 5-methyl-1H-pyrazol-4-yl)-6-(tert-butoxycarbonyl)pyridin-2-yl)- 5-(2-(tert-butyldiphenylsilyl)ethoxy)-1,2,3,4- tetrahydroisoquinoline-8-carboxylic acid
Example 1.5.12 (1.15 g) was dissolved in tetrahydrofuran (4.5 mL), and methanol (2.2 mL), water (2.2 mL), and lithium hydroxide monohydrate (96 mg) were added. The reaction mixture was stirred at room temperature for five days. Water (20 mL) and 2N aqueous HCl (1.1 mL) were added. The mixture was extracted with ethyl acetate, and the organic layer was washed with brine and dried over Na2SO4. After filtration and concentration, purification by silica gel chromatography, eluting with dichloromethane/ethyl acetate (70/30) followed by dichloromethane/ethyl acetate/acetic acid (70/30/1), gave the title compound.
1.5.14. tert-butyl 3-(1-((3-(2-azidoethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-5-(2-(tert-butyldiphenylsilyl)ethoxy)-3,4- dihydroisoquinolin-2(1H)-yl)picolinate
Example 1.5.13 (80 mg) and benzo[d]thiazol-2-amine (14 mg) were dissolved in
dichloromethane (1.2 mL). N,N-Dimethylpyridin-4-amine (17 mg) and N-ethyl-N’-(3- dimethylaminopropyl)carbodiimide hydrochloride (27 mg) were added and the reaction was stirred at room temperature overnight. The reaction was concentrated and the crude residue was purified by silica gel chromatography, eluting with dichloromethane/ethyl acetate (90/10), to provide the title compound MS (ESI) m/e 11103 (M+H)+ 1.5.15. tert-butyl 3-(1-((3-(2-azidoethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-5-hydroxy-3,4-dihydroisoquinolin-2(1H)- yl)picolinate
Example 1.5.14 (160 mg) was dissolved in a 1.0M solution of tetrabutylammonium fluoride in 95/5 tetrahydrofuran/water (1.15 mL) and the reaction was heated at 60 °C for two days. Powdered 4Å molecular sieves were added, and the mixture was heated at 60 °C for another day. The reaction was cooled, then concentrated and the crude residue was purified by silica gel chromatography, eluting with 70/30/1 dichloromethane/ethyl acetate/acetic acid, to provide the title compound. MS (ESI) m/e 844.2 (M+H)+.
1.5.16. tert-butyl 3-(1-((3-(2-aminoethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-5-hydroxy-3,4-dihydroisoquinolin-2(1H)- yl)picolinate
Example 1.5.15 (70 mg) was dissolved in tetrahydrofuran (2 mL), 10% palladium on carbon (20 mg) was added, and the mixture was stirred under a hydrogen balloon overnight. After filtration through diatomaceous earth and evaporation of the solvent, the crude title compound was purified by reverse phase chromatography (C18 column), eluting with 10-90% acetonitrile in 0.1% TFA water, to provide the title compound as a trifluoroacetic acid salt.
1.5.17. 3-(1-((3-(2-aminoethoxy)-5,7-dimethyladamantan-1-yl)methyl)-5- methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5- hydroxy-3,4-dihydroisoquinolin-2(1H)-yl)picolinic acid
Example 1.5.16 (11 mg) was dissolved in 4N HCl in dioxane (0.5 mL) and stirred at room temperature overnight. The solids were filtered off and washed with dioxane to provide the title compound as a hydrochloride salt. 1H NMR (500 MHz, dimethyl sulfoxide-d6) δ ppm 12.60 (v br s, 1H), 10.40 (br s, 1H), 8.00 (d, 1H) 7.76 (d, 1H), 7.75 (br s, 3H), 7.60 ( d, 1H), 7.51 (d, 1H), 7.46 (t, 1H), 7.33 (t, 1H), 7.30 (s, 1H), 6.98 (d, 1H), 6.82 (d, 1H), 4.99 (s, 2H), 3.89 (m, 2H), 3.83 (s, 2H), 3.50 (m, 2H), 2.88 (m, 2H), 2.79 (m, 2H), 2.11 (s, 3H), 1.41 (s, 2H), 1.29 (m, 4H), 1.14 (m, 4H), 1.04 (m, 2H), 0.87 (s, 6H). MS (ESI) m/e 762.2 (M+H)+.
1.6. Synthesis of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-3-[1- ({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid (Compound W3.06)
1.6.1. tert-butyl 3-(1-((3-(2-((tert-butoxycarbonyl)(methyl)amino) ethoxy)-5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H- pyrazol-4-yl)-6-(8-(methoxycarbonyl)naphthalen-2-yl)picolinate To a solution of methyl 7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-naphthoate (2.47 g) in dioxane (40 mL) and water (20 mL) was added Example 1.1.11 (4.2 g),
bis(triphenylphosphine)palladium(II) dichloride (556 mg), and CsF (3.61 g). The mixture was stirred at reflux overnight. The mixture was diluted with ethyl acetate (400 mL) and washed with water and brine, and dried over Na2SO4. After filtration and evaporation of the solvent, the crude material was purified via column chromatography, eluting with 20% ethyl acetate in heptane followed by 5% methanol in dichloromethane, to provide the title compound. MS (ESI) m/e 793.4 (M+H)+.
1.6.2. 7-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert-butoxycarbonyl)
(methyl)amino)ethoxy)-5,7-dimethyladamantan-1-yl)methyl)-5- methyl-1H-pyrazol-4-yl)pyridin-2-yl)-1-naphthoic acid
To a solution of Example 1.6.1 (500 mg) in tetrahydrofuran (4 mL), methanol (2 mL) and water (2 mL) was added lithium hydroxide monohydrate (500 mg). The mixture was stirred for 3 hours. The mixture was then acidified with 1N aqueous HCl and diluted with ethyl acetate (200 mL). The organic layer was washed with water and brine, and dried over Na2SO4. Filtration and evaporation of the solvent gave the crude title compound which was used in the next reaction without further purification. MS (ESI) m/e 779.4 (M+H)+.
1.6.3. 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-3-[1-({3,5- dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid
To a solution of Example 1.6.2 (79 mg) in N,N-dimethylformamide (2 mL) was added benzo[d]thiazol-2-amine (23 mg), fluoro-N,N,N',N'-tetramethylformamidinium hexafluorophosphate (41 mg) and N,N-diisopropylethylamine (150 mg). The mixture was stirred at 60 °C for 3 hours. The reaction mixture was diluted with ethyl acetate (200 mL) and washed with water and brine, and dried over Na2SO4. Filtration and evaporation of the solvent gave a crude intermediate which was dissolved in dichloromethane/TFA (1:1, 6 mL) and left to sit overnight. Evaporation of the solvent gave a residue which was dissolved in dimethyl sulfoxide/methanol (1:1, 9 mL) and purified by HPLC (Gilson system, eluting with 10-85% acetonitrile in 0.1% TFA in water) to give the pure title compound. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ ppm 13.11 (s, 1H), 9.02 (s, 1H), 8.38 (dd, 1H), 8.26-8.34 (m, 2H), 8.13-8.27 (m, 3H), 8.07 (d, 1H), 8.02 (d, 1H), 7.93 (d, 1H), , 7.82 (d, 1H), 7.67-7.75 (m, 1H), , 7.44-7.53 (m, 2H), 7.30-7.41 (m, 1H), 3.90 (s, 3H), 2.94-3.12 (m, 3H), 2.53-2.60 (m, 4H), 2.20-2.31 (m, 3H), 1.45 (s, 2H), 1.25-1.39 (m, 4H), 0.99-1.23 (m, 4H), 0.89 (s, 6 H). MS (ESI) m/e 755.4 (M+H)+.
1.7. Synthesis of 3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H- pyrazol-4-yl]-6-[8-([1,3]thiazolo[5,4-b]pyridin-2- ylcarbamoyl)naphthalen-2-yl]pyridine-2-carboxylic acid (Compound
W307) The title compound was prepared by substituting thiazolo[5,4-b]pyridin-2-amine for benzo[d]thiazol-2-amine in Example 1.6.3. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 13.25 (s, 1H), 9.02 (s, 1H), , 8.54 (dd, 1H), 8.39 (dd, 1H), 8.14-8.35 (m, 6H), 8.04 (d, 1H), 7.93 (d, 1H), 7.66-7.75 (m, 1H), 7.55 (dd, 1H), 7.49 (s, 1H), 3.57 (t, 3H), 2.95-3.10 (m, 2H), 2.51-2.62 (m, 3H), 2.19-2.28 (m, 3H), 1.45 (s, 2H), 1.24-1.38 (m, 4H), 0.98-1.24 (m, 6H), 0.89 (s, 6 H). MS (ESI) m/e 756.3 (M+H)+.
1.8. Synthesis of 3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H- pyrazol-4-yl]-6-[8-([1,3]thiazolo[4,5-b]pyridin-2- ylcarbamoyl)naphthalen-2-yl]pyridine-2-carboxylic acid (Compound
W3.08)
The title compound was prepared by substituting thiazolo[4,5-c]pyridin-2-amine for benzo[d]thiazol-2-amine in Example 1.6.3. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ ppm 13.40 (s, 1H), 9.04 (s, 1H), 8.62 (dd, 1H), 8.56 (dd, 1H), 8.39 (dd, 1H), 8.13-8.34 (m, 5H), 8.06 (d, 1H), 7.94 (d, 1H), 7.68-7.79 (m, 1H), 7.45-7.54 (m, 1H), 7.39 (dd, 1H), 3.90 (s, 3H), 3.54-3.60 (m, 3H), 2.94-3.08 (m, 2H), 2.51-2.60 (m, 4H), 2.18-2.31 (m, 3H), 1.46 (s, 2H), 1.24-1.40 (m, 4H), 1.01-1.21 (m, 6H), 0.83-0.89 (m, 5 H). MS (ESI) m/e 756.3 (M+H)+.
1.9. Synthesis of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H- pyrazol-4-yl]pyridine-2-carboxylic acid (Compound W3.09)
1.9.1. tert-butyl 8-bromo-5-hydroxy-3,4-dihydroisoquinoline-2(1H)- carboxylate
To a solution of tert-butyl 5-hydroxy-3,4-dihydroisoquinoline-2(1H)-carboxylate (9 g) in N,N-dimethylformamide (150 mL) was added N-bromosuccinimide (6.43 g). The mixture was stirred overnight and quenched with water (200 mL). The mixture was diluted with ethyl acetate (500 mL) and washed with water and brine, and dried over sodium sulfate. Filtration and evaporation of the solvent gave crude title compound which was used in the next reaction without further purification. MS(ESI) m/e 329.2 (M+H)+.
1.9.2. tert-butyl 5-(benzyloxy)-8-bromo-3,4-dihydroisoquinoline-2(1H)- carboxylate
To a solution of Example 1.9.1 (11.8 g) in acetone (200 mL) was added benzyl bromide (7.42 g) and K2CO3 (5 g). The mixture was stirred at reflux overnight. The mixture was concentrated and the residue was partitioned between ethyl acetate (600 mL) and water (200 mL). The organic layer was washed with water and brine, and dried over sodium sulfate. Filtration and evaporation of the solvent gave crude title compound which was purified on a silica gel column and eluted with 10% ethyl acetate in heptane to provide the title compound MS (ESI) m/e 4181 (M+H)+ 1.9.3. 2-tert-butyl 8-methyl 5-(benzyloxy)-3,4-dihydroisoquinoline- 2,8(1H)-dicarboxylate
Methanol (100 mL) and triethylamine (9.15 mL) were added to Example 1.9.2 (10.8 g) and [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (0.48 g) in a 500 mL stainless steel pressure reactor. The vessel was sparged with argon several times. The reactor was pressurized with carbon monoxide and stirred for 2 hours at 100 °C under 60 psi of carbon monoxide. After cooling, the crude reaction mixture was concentrated under vacuum. The residue was partitioned between ethyl acetate (500 mL) and water (200 mL). The organic layer was further washed with water and brine, and dried over sodium sulfate. After filtration and evaporation of the solvent, the residue was purified on a 330g silica gel column, eluting with 10-20% ethyl acetate in heptane, to provide the title compound. MS (ESI) m/e 398.1 (M+H)+.
1.9.4. methyl 5-(benzyloxy)-1,2,3,4-tetrahydroisoquinoline-8- carboxylate hydrochloride
To a solution of Example 1.9.3 (3.78 g) in tetrahydrofuran (20 mL) was added 4N HCl in dioxane (20 mL). The mixture was stirred overnight and the mixture was concentrated under vacuum and the crude title compound was used in the next reaction without further purification. MS (ESI) m/e 298.1 (M+H)+.
1.9.5. methyl 5-(benzyloxy)-2-(5-bromo-6-(tert- butoxycarbonyl)pyridin-2-yl)-1,2,3,4-tetrahydroisoquinoline-8- carboxylate
To a solution of Example 1.9.4 (3.03 g) in dimethyl sulfoxide (50 mL) was added Example 1.4.4 (2.52 g) and triethylamine (3.8 mL). The mixture was stirred at 60 °C overnight under nitrogen. The reaction mixture was diluted with ethyl acetate (500 mL) and washed with water and brine, and dried over sodium sulfate. After filtration and evaporation of the solvent, the crude material was purified on a silica gel column, eluting with 20% ethyl acetate in heptane, to give the title compound. MS (ESI) m/e 553.1 (M+H)+.
1.9.6. methyl 5-(benzyloxy)-2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2- ((tert-butoxycarbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)pyridin-2-yl)-1,2,3,4-tetrahydroisoquinoline-8-carboxylate
To a solution of Example 1.9.5 (2.58 g) in tetrahydrofuran (40 mL) and water (20 mL) was added Example 1.1.10 (2.66 g), 1,3,5,7-tetramethyl-6-phenyl--2,4,8-trioxa--6-phosphaadamante (341 mg), tris(dibenzylideneacetone)dipalladium(0) (214 mg), and K3PO4 (4.95 g). The mixture was stirred at reflux for 4 hours. The mixture was diluted with ethyl acetate (500 mL) and washed with water and brine, and dried over sodium sulfate. After filtration and evaporation of the solvent, the crude material was purified on a silica gel column, eluting with 20% ethyl acetate in dichloromethane, to give the title compound MS (ESI) m/e 9045 (M+H)+ 1.9.7. methyl 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan- 1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-5-hydroxy- 1,2,3,4-tetrahydroisoquinoline-8-carboxylate
Example 1.9.6 (3.0 g) in tetrahydrofuran (60 mL) was added to Pd(OH)2 (0.6 g, Degussa #E101NE/W, 20% on carbon, 49% water content) in a 250 mL SS pressure bottle. The mixture was agitated for 16 hours under 30 psi of hydrogen gas at 50 °C. The mixture was then filtered through a nylon membrane, and the solvent concentrated under vacuum to provide the title compound. MS (ESI) m/e 815.1(M+H)+.
1.9.8. methyl 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan- 1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-5-methoxy- 1,2,3,4-tetrahydroisoquinoline-8-carboxylate
Example 1.9.7 (170 mg) was dissolved in dichloromethane (0.8 mL) and methanol (0.2 mL). To the mixture was added a 2.0M solution of (trimethylsilyl)diazomethane in diethyl ether (0.17 mL) and the reaction was stirred at room temperature overnight. Additional 2.0M
(trimethylsilyl)diazomethane in diethyl ether (0.10 mL) was added, and the reaction was allowed to stir for 24 hours. The reaction mixture was then concentrated and the title compound was used without further purification. MS (ESI) m/e 828.2 (M+H)+.
1.9.9. 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan- 1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-5-methoxy- 1,2,3,4-tetrahydroisoquinoline-8-carboxylic acid
The title compound was prepared by substituting Example 1.9.8 for Example 1.5.12 in Example 1.5.13. MS (ESI) m/e 814.1 (M+H)+.
1.9.10. tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan- 1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate
The title compound was prepared by substituting Example 1.9.9 for Example 1.5.13 in Example 1.5.14. MS (ESI) m/e 946.1 (M+H)+.
1.9.11. 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3,5-dimethyl-7-(2- (methylamino)ethoxy)adamantan-1-yl)methyl)-5-methyl-1H- pyrazol-4-yl)picolinic acid
The title compound was prepared by substituting Example 1.9.10 for Example 1.5.16 in Example 1517 1H NMR (500 MHz dimethyl sulfoxide-d6) δ ppm 874 (br s 2H) 802 (d 1H) 777 (m, 2H), 7.54 (d, 1H), 7.47 (t, 1H), 7.34 (m, 2H), 7.01 (d, 2H), 5.01 (s, 2H), 3.90 (m, 2H), 3.89 (s, 3H), 3.85 (s, 2H), 3.58 (m, 2H), 3.57 (s, 3H), 2.98 (m, 2H), 2.82 (m, 2H), 2.12 (s, 3H), 1.41 (s, 2H), 1.30 (m, 4H), 1.14 (m, 4H), 1.04 (m, 2H), 0.87 (s, 6H). MS (ESI) m/e 790.2 (M+H)+.
1.10. Synthesis of 6-[5-(1,3-benzothiazol-2-ylcarbamoyl)quinolin-3-yl]-3-[1- ({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid (Compound W3.10)
1.10.1. 3-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan- 1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)quinoline-5- carboxylic acid
A mixture of 3-bromoquinoline-5-carboxylic acid (300 mg), 4,4,4',4',5,5,5',5'-octamethyl-2,2'- bi(1,3,2-dioxaborolane) (363 mg), and potassium acetate (350 mg) in dioxane (5 mL) was purged with nitrogen gas for 5 minutes, and PdCl2(dppf)-CH2Cl2 adduct (58.3 mg) was added. The mixture was heated at 100 °C overnight and cooled. To this mixture was added Example 1.1.11 (510 mg), dichlorobis(triphenylphosphine)-palladium(II) (83 mg), CsF (362 mg), and water (3 mL). The resulting mixture was heated at 100 °C overnight and filtered through diatomaceous earth. The filtrate was concentrated, and the residue was dissolved in dimethyl sulfoxide, loaded onto a C18 column (300g), and eluted with a gradient of 50-100% acetonitrile in a 0.1% TFA/water solution to provide the title compound. MS (ESI) m/e 780.5 (M+H)+.
1.10.2. tert-butyl 6-(5-(benzo[d]thiazol-2-ylcarbamoyl)quinolin-3-yl)-3- (1-((3-(2-((tert-butoxycarbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)picolinate
To a mixture of Example 1.10.1 (120 mg), benzo[d]thiazol-2-amine (46.2 mg), and O-(7- azabenzotriazol-1-yl)-N,N,N’,N’-tetramethyluronium hexafluorophosphate (HATU, 117 mg) in N,N- dimethylformamide (0.5 mL) was added N,N-diisopropylethylamine (134 µL). The mixture was stirred overnight and loaded onto a C18 column (300 g), eluting with a gradient of 50-100% acetonitrile in 0.1% TFA/water solution to provide the title compound. MS (ESI) m/e 913.4 (M+H)+.
1.10.3. 6-[5-(1,3-benzothiazol-2-ylcarbamoyl)quinolin-3-yl]-3-[1-({3,5- dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid
Example 1.10.2 (50 mg) in dichloromethane (3 mL) was treated with trifluoroacetic acid (2 mL) overnight and concentrated. The residue was dissolved in a mixture of dimethyl sulfoxide (5 mL), loaded onto a C18 column (300 g), and eluted with a gradient of 10-70% acetonitrile in 0.1% TFA water solution to provide the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 1322 (s 1H) 973 (d 1H) 941 (s 1H) 834 (dd 2H) 827 (s 3H) 818 (d 1H) 808 (d 1H) 802- 7.93 (m, 2H), 7.82 (d, 1H), 7.55-7.46 (m, 2H), 7.38 (t, 1H), 3.91 (s, 2H), 3.03 (p, 2H), 2.59-2.53 (m, 4H), 2.25 (s, 3H), 1.46 (s, 2H), 1.38-1.25 (m, 4H), 1.18 (s, 4H), 1.11-1.01 (m, 2H), 0.89 (s, 6H). MS (ESI) m/e 756.2 (M+H)+.
1.11. Synthesis of 6-[4-(1,3-benzothiazol-2-ylcarbamoyl)quinolin-6-yl]-3-[1- ({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid (Compound W3.11)
1.11.1. ethyl 6-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan- 1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)quinoline-4- carboxylate
The title compound was prepared as described in Example 1.10.1, replacing 3- bromoquinoline-5-carboxylic acid with ethyl 6-bromoquinoline-4-carboxylate. MS (ESI) m/e 808.4 (M+H)+.
1.11.2. 6-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan- 1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)quinoline-4- carboxylic acid
To a solution of Example 1.11.1 (100 mg) in dimethyl sulfoxide (2 mL) was added methanol (2 mL) and 1M lithium hydroxide (248 µL). The mixture was stirred for 30 minutes, acidified to pH 4 with 10% HCl, diluted with ethyl acetate and washed with water and brine to provide the title compound. MS (ESI) m/e 780.4 (M+H)+.
1.11.3. tert-butyl 6-(4-(benzo[d]thiazol-2-ylcarbamoyl)quinolin-6-yl)-3- (1-((3-(2-((tert-butoxycarbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)picolinate
The title compound was prepared as described in Example 1.10.2, replacing Example 1.10.1 with Example 1.11.2. MS (ESI) m/e 912.3 (M+H)+.
1.11.4. 6-[4-(1,3-benzothiazol-2-ylcarbamoyl)quinolin-6-yl]-3-[1-({3,5- dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid
The title compound was prepared as described in Example 1.10.3, replacing Example 1.10.2 with Example 1.11.3. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 13.34 (s, 2H), 9.14 (d, 1H), 8.94 (s, 1H), 8.63 (dd, 1H), 8.27 (dd, 4H), 8.09 (d, 1H), 8.00-7.90 (m, 2H), 7.83 (d, 1H), 7.50 (d, 2H), 7.40 (t, 1H), 3.90 (s, 2H), 3.03 (p, 2H), 2.56 (t, 4H), 2.23 (s, 3H), 1.45 (s, 2H), 1.32 (d, 3H), 1.18 (s, 4H), 1.11-0.98 (m, 2H), 0.89 (s, 6H). MS (ESI) m/e 756.2 (M+H)+. 1.12. Synthesis of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-3-{1-[(3-{2-[(2- methoxyethyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl)methyl]-5-methyl-1H-pyrazol-4-yl}pyridine-2-carboxylic acid (Compound W3.12)
1.12.1. methyl 5-(benzyloxy)-2-(6-(tert-butoxycarbonyl)-5-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-2-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxylate
The title compound was prepared by substituting Example 1.9.5 for Example 1.5.9 in Example 1.5.11. MS (DCI) m/e 601.0 (M+H)+.
1.12.2. 2-((3-((4-iodo-5-methyl-1H-pyrazol-1-yl)methyl)-5,7- dimethyladamantan-1-yl)oxy)acetaldehyde
Dimethyl sulfoxide (4.8 mL) was dissolved in dichloromethane (150 mL). The mixture was cooled to -75 °C, and oxalyl chloride (2.6 mL) was added dropwise. The reaction mixture was stirred at -75 °C for 45 minutes, and a solution of Example 1.1.6 (7.1 g) in dichloromethane (45 mL) was added dropwise. The reaction mixture was stirred at -75 °C for 30 minutes, and triethylamine (5.0 mL) was added. The reaction was warmed to room temperature, poured into water, and extracted with diethyl ether. The organic layer was washed with brine and dried over Na2SO4. After filtration and concentration, purification by silica gel chromatography, eluting with dichloromethane/ethyl acetate 85/15, gave the title compound. MS (DCI) m/e 443.0 (M+H)+.
1.12.3. 2-((3-((4-iodo-5-methyl-1H-pyrazol-1-yl)methyl)-5,7- dimethyladamantan-1-yl)oxy)-N-(2-methoxyethyl)ethanamine
Example 1.12.2 (4.0 g) and 2-methoxyethanamine (0.90 mL) were dissolved in
dichloromethane (40 mL) and the mixture was stirred at room temperature for two hours. A suspension of sodium borohydride (500 mg) in methanol (7 mL) was added and the resulting mixture was stirred for 45 minutes. The reaction was then added to saturated aqueous NaHCO3 and resultant mixture extracted with ethyl acetate. The organic layer was washed with brine and dried over Na2SO4. The title compound was obtained after filtration and concentration and was used without purification. MS (DCI) m/e 502.1 (M+H)+.
1.12.4. tert-butyl (2-((3-((4-iodo-5-methyl-1H-pyrazol-1-yl)methyl)-5,7- dimethyladamantan-1-yl)oxy)ethyl)(2-methoxyethyl)carbamate
Example 1.12.3 (4.4 g) was dissolved in tetrahydrofuran (60 mL), and di-tert-butyl dicarbonate (3.0 g) and N,N-dimethylpyridin-4-amine (0.15 g) were added. The reaction was stirred at room temperature overnight. The reaction was then concentrated and purified by flash chromatography, eluting with dichloromethane/ethyl acetate (3/1), to provide the title compound. 1.12.5. methyl 5-(benzyloxy)-2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2- ((tert-butoxycarbonyl)(2-methoxyethyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)pyridin-2-yl)-1,2,3,4-tetrahydroisoquinoline-8-carboxylate
The title compound was prepared by substituting Example 1.12.1 for Example 1.5.11 and Example 1.12.4 for Example 1.5.10 in Example 1.5.12. MS (ESI) m/e 948.2 (M+H)+.
1.12.6. methyl 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(2-methoxyethyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)pyridin-2-yl)-5-hydroxy-1,2,3,4-tetrahydroisoquinoline-8- carboxylate
Example 1.12.5 (5.2 g) was dissolved in tetrahydrofuran (100 mL).20% Palladium hydroxide on activated charcoal (1.0 g) was then added, and the reaction mixture agitated on a Parr rector under a hydrogen atmosphere at 30 psi and 50 °C for 3 hours. After filtration and concentration, purification by silica gel chromatography, eluting with heptanes/ethyl acetate (2/3), gave the title compound. MS (ESI) m/e 858.1 (M+H)+.
1.12.7. methyl 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(2-methoxyethyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)pyridin-2-yl)-5-methoxy-1,2,3,4-tetrahydroisoquinoline-8- carboxylate
The title compound was prepared by substituting Example 1.12.6 for Example 1.9.7 in Example 1.9.8. MS (ESI) m/e 872.2 (M+H)+.
1.12.8. 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert-butoxycarbonyl)(2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-5-methoxy- 1,2,3,4-tetrahydroisoquinoline-8-carboxylic acid
The title compound was prepared by substituting Example 1.12.7 for Example 1.5.12 in Example 1.5.13. MS (ESI) m/e 858.1 (M+H)+.
1.12.9. tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-((tert- butoxycarbonyl)(2-methoxyethyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)picolinate
The title compound was prepared by substituting Example 1.12.8 for Example 1.5.13 in Example 1.5.14. MS (ESI) m/e 990.1 (M+H)+. 1.12.10. 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-(((1r,3s,5R,7S)-3-(2-((2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinic acid
Example 1.12.9 (2.6 g) was dissolved in dioxane (20 mL), then 4N HCl in dioxane (100 mL) was added, and the reaction was stirred at room temperature overnight. The precipitants were allowed to settle and the supernatant was drawn off. The remaining solids were purified by reverse phase chromatography (C18 column), eluting with 10-90% acetonitrile in 0.1% TFA/water, to provide the title compound as a trifluoroacetic acid salt. 1H NMR (500 MHz, dimethyl sulfoxide-d6) δ ppm 8.41 (v br s, 2H), 8.01 (d, 1H) 7.77 (m, 2H), 7.50 (d, 1H), 7.47 (m, 1H), 7.34 (t, 1H), 7.29 (s, 1H), 7.01 (dd, 2H), 5.00 (s, 2H), 3.90 (m, 2H), 3.89 (s, 3H), 3.83 (s, 2H), 3.56 (m, 4H), 3.29 (s, 3H), 3.12 (m, 2H), 3.05 (m, 2H), 2.81 (m, 2H), 2.11 (s, 3H), 1.41 (s, 2H), 1.30 (m, 4H), 1.14 (m, 4H), 1.04 (m, 2H), 0.87 (s, 6H). MS (ESI) m/e 834.3 (M+H)+.
1.13. Synthesis of 3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec- 1-yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-5-cyano-3,4-dihydroisoquinolin-2(1H)-yl]pyridine-2- carboxylic acid (Compound W3.13)
1.13.1. 4-Bromo-3-cyanomethyl-benzoic acid methyl ester
Trimethylsilanecarbonitrile (3.59 mL) was added to tetrahydrofuran (6 mL). 1M
Tetrabutylammonium fluoride (26.8 mL) was added dropwise over 30 minutes. The solution was then stirred at room temperature for 30 minutes. Methyl 4-bromo-3-(bromomethyl)benzoate (7.50 g) was dissolved in acetonitrile (30 mL) and the resultant solution added to the first solution dropwise over 30 minutes. The solution was then heated to 80 °C for 30 minutes and then allowed to cool to room temperature. The solution was concentrated under reduced pressure and purified by flash column chromatography on silica gel, eluting with 20-30% ethyl acetate in heptanes. The solvent was evaporated under reduced pressure to provide the title compound.
1.13.2. 3-(2-Aminoethyl)-4-bromobenzoic acid methyl ester
Example 1.13.1 (5.69 g) was dissolved in tetrahydrofuran (135 mL), and 1 M borane (in tetrahydrofuran, 24.6 mL) was added. The solution was stirred at room temperature for 16 hours and then slowly quenched with methanol and 1M HCL. 4M HCl (150 mL) was added, and the solution was stirred at room temperature for 16 hours. The mixture was concentrated was reduced under reduced pressure, and the pH adjusted to between 11 and 12 using solid potassium carbonate. The solution was then extracted with dichloromethane (3x 100 mL). The organic extracts were combined and dried over anhydrous sodium sulfate. The solution was filtered and concentrated under reduced pressure, and the material was purified by flash column chromatography on silica gel, eluting with 10- 20% methanol in dichloromethane. The solvent was evaporated under reduced pressure to provide the title compound MS (ESI) m/e 258 260 (M+H)+ 1.13.3. 4-Bromo-3-[2-(2,2,2-trifluoroacetylamino)-ethyl]-benzoic acid
methyl ester
Example 1.13.2 (3.21 g) was dissolved in dichloromethane (60 mL). The solution was cooled to 0 °C, and triethylamine (2.1 mL) was added. Trifluoroacetic anhydride (2.6 mL) was then added dropwise. The solution was stirred at 0 °C for ten minutes and then allowed to warm to room temperature while stirring for one hour. Water (50 mL) was added and the solution was diluted with ethyl acetate (100 mL). 1M HCl was added (50 mL) and the organic layer was separated, washed with 1M HCl, and then washed with brine. The organic layer was then dried on anhydrous sodium sulfate. After filtration, the solvent was evaporated under reduced pressure to provide the title compound. MS (ESI) m/e 371, 373 (M+H)+.
1.13.4. 5-Bromo-2-(2,2,2-trifluoroacetyl)-1,2,3,4-tetrahydroisoquinoline- 8-carboxylic acid methyl ester
Example 1.13.3 (4.40 g) and paraformaldehyde (1.865 g) were placed in a flask and concentrated sulfuric acid (32 mL) was added. The solution was stirred at room temperature for one hour. Cold water (120 mL) was added. The solution was extracted with ethyl acetate (3x 100 mL). The extracts were combined, washed with saturated aqueous sodium bicarbonate (100 mL), washed with water (100 mL), and dried over anhydrous sodium sulfate. The solution was concentrated under reduced pressure, and the material was purified by flash column chromatography on silica gel, eluting with 20-30% ethyl acetate in heptanes. The solvent was evaporated under reduced pressure to provide the title compound. MS (ESI) m/e 366, 368 (M+H)+.
1.13.5. 5-Cyano-2-(2,2,2-trifluoroacetyl)-1,2,3,4-tetrahydroisoquinoline- 8-carboxylic acid methyl ester
Example 1.13.4 (500 mg) and dicyanozinc (88 mg) were added to N,N-dimethylformamide (4 mL). The solution was degassed and flushed with nitrogen three times.
Tetrakis(triphenylphosphine)palladium(0) (79 mg) was added, and the solution was degassed and flushed with nitrogen once. The solution was then stirred at 80 °C for 16 hours. The solution was cooled, diluted with 50% ethyl acetate in heptanes (20 mL), and washed with 1 M hydrochloric acid (15 mL) twice. The organic layer was washed with brine and dried over anhydrous sodium sulfate. The solution was filtered and concentrated under reduced pressure, and the material was purified by flash column chromatography on silica gel, eluting with 20-30% ethyl acetate in heptanes. The solvent was evaporated under reduced pressure to provide the title compound.
1.13.6. 5-Cyano-1,2,3,4-tetrahydroisoquinoline-8-carboxylic acid methyl
ester
Example 1.13.5 (2.00 g) was dissolved in methanol (18 mL) and tetrahydrofuran (18 mL). Water (9 mL) was added followed by potassium carbonate (1.064 g). The reaction was stirred at room temperature for 135 minutes and then diluted with ethyl acetate (100 mL). The solution was washed with saturated aqueous sodium bicarbonate and dried on anhydrous sodium sulfate The solvent was filtered and evaporated under reduced pressure to provide the title compound. MS (ESI) m/e 217 (M+H)+.
1.13.7. 2-(5-Bromo-6-tert-butoxycarbonylpyridin-2-yl)-5-cyano-1,2,3,4- tetrahydroisoquinoline-8-carboxylic acid methyl ester
Example 1.13.6 (1.424 g) and Example 1.4.4 (1.827 g) were dissolved in dimethyl sulfoxide (13 mL). N,N-Diisopropylethylamine (1.73 mL) was added, and the solution was heated to 50 °C for 16 hours. Additional Example 1.4.4 (0.600 g) was added, and the solution was heated at 50 °C for another 16 hours. The solution was allowed to cool to room temperature, diluted with ethyl acetate (50 mL), washed with water (25 mL) twice, washed with brine, and then dried on anhydrous sodium sulfate. The solution was filtered and concentrated under reduced pressure, and the material was purified by flash column chromatography on silica gel, eluting with 20-50% ethyl acetate in heptanes. The solvent was evaporated under reduced pressure to provide the title compound. MS (ESI) m/e 472, 474 (M+H)+.
1.13.8. 2-[6-tert-Butoxycarbonyl-5-(4,4,5,5-tetramethyl- [1,3,2]dioxaborolan-2-yl)-pyridin-2-yl]-5-cyano-1,2,3,4- tetrahydroisoquinoline-8-carboxylic acid methyl ester
Example 1.13.7 (2.267 g) and triethylamine (1.34 mL) were added to acetonitrile (15 mL). The solution was degassed and flushed with nitrogen three times. 4,4,5,5-Tetramethyl-1,3,2- dioxaborolane (1.05 mL) was added followed by dichloro[1,1’- bis(diphenylphosphino)ferrocene]palladium(II) (196 mg). The solution was degassed and flushed with nitrogen once and heated to reflux for 16 hours. The solution was cooled, diluted with ethyl acetate (50 mL), washed with water (10 mL), washed with brine, and dried on anhydrous sodium sulfate. The solution was concentrated under reduced pressure, and the material was purified by flash column chromatography on silica gel, eluting with 20-30% ethyl acetate in heptanes. The solvent was evaporated under reduced pressure to provide the title compound. MS (ESI) m/e 520 (M+H)+.
1.13.9. 2-(6-tert-Butoxycarbonyl-5-{1-[5-(2-tert-butoxycarbonylamino- ethoxy)-3,7-dimethyl-adamantan-1-ylmethyl]-5-methyl-1H- pyrazol-4-yl}-pyridin-2-yl)-5-cyano-1,2,3,4-tetrahydro- isoquinoline-8-carboxylic acid methyl ester
Example 1.13.8 (140 mg) and Example 1.4.2 (146 mg) were dissolved in tetrahydrofuran (3 mL). Potassium phosphate (286 mg) and water (0.85 mL) were added. The solution was degassed and flushed with nitrogen three times. (1S,3R,5R,7S)-1,3,5,7-Tetramethyl-8-tetradecyl-2,4,6-trioxa- 8-phosphaadamantane (11 mg) and tris(dibenzylideneacetone)dipalladium(0) (12 mg) were added, and the solution was degassed and flushed with nitrogen once. The solution was heated to 62 °C for 16 hours. The solution was cooled, then diluted with water (5 mL) and ethyl acetate (25 mL). The organic layer was separated and washed with brine and dried on anhydrous sodium sulfate. The solution was filtered and concentrated under reduced pressure and the material was purified by flash column chromatography on silica gel, eluting with 30-50% ethyl acetate in heptanes. The solvent was evaporated under reduced pressure to provide the title compound. MS (ESI) m/e 809 (M+H)+.
1.13.10. 2-(6-tert-Butoxycarbonyl-5-{1-[5-(2-tert-butoxycarbonylamino- ethoxy)-3,7-dimethyl-adamantan-1-ylmethyl]-5-methyl-1H- pyrazol-4-yl}-pyridin-2-yl)-5-cyano-1,2,3,4-tetrahydro- isoquinoline-8-carboxylic acid
Example 1.13.9 (114 mg) was dissolved in tetrahydrofuran (0.7 mL) and methanol (0.35 mL). Water (0.35 mL) was added followed by lithium hydroxide monohydrate (11 mg). The solution was stirred at room temperature for 16 hours, and 1 M hydrochloric acid (0.27 mL) was added. Water (1 mL) was added and the solution was extracted with ethyl acetate (5 mL) three times. The extracts were combined and dried on anhydrous sodium sulfate and filtered. The solvent was evaporated under reduced pressure to provide the title compound. MS (ESI) m/e 795 (M+H)+.
1.13.11. 6-[8-(Benzothiazol-2-ylcarbamoyl)-5-cyano-3,4-dihydro-1H- isoquinolin-2-yl]-3-{1-[5-(2-tert-butoxycarbonylamino-ethoxy)- 3,7-dimethyl-adamantan-1-ylmethyl]-5-methyl-1H-pyrazol-4-yl}- pyridine-2-carboxylic acid tert-butyl ester
Example 1.13.10 (89 mg) and benzo[d]thiazol-2-amine (18 mg) were dissolved in dichloromethane (1.2 mL). N-(3-Dimethylaminopropyl)-N’-ethylcarbodiimide hydrochloride (39 mg) and N,N-dimethylpyridin-4-amine (25 mg) were added, and the solution was stirred at room temperature for 16 hours. The material was purified by flash column chromatography on silica gel, eluting with 50% ethyl acetate in heptanes. The solvent was evaporated under reduced pressure to provide the title compound. MS (ESI) m/e 927 (M+H)+.
1.13.12. 3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-5-cyano-3,4-dihydroisoquinolin-2(1H)-yl]pyridine- 2-carboxylic acid
Example 1.13.11 (44 mg) was dissolved in dichloromethane (1 mL). Trifluoroacetic acid (0.144 mL) was added and the solution stirred at room temperature for 16 hours. The solvents were then evaporated under reduced pressure, the residue was dissolved in dichloromethane (1 mL), and the solvent removed under reduced pressure. Diethyl ether was added (2 mL) and was removed under reduced pressure. Diethyl ether (2 mL) was added again and removed under reduced pressure to provide the title compound as the trifluoroacetic acid salt. 1H NMR (400MHz, dimethyl sulfoxide-d6) δ ppm 8.52 (bs, 1H), 8.05 (d, 1H), 7.92 (d, 1H), 7.82-7.75 (m, 2H), 7.63 (m, 2H), 7.50 (dd, 2H), 7.42- 7.28 (m, 3H), 7.16 (t, 1H), 7.04 (d, 1H), 4.98 (s, 2H), 3.96 (t, 2H), 3.83 (s, 2H), 3.49 (t, 2H), 3.15 (t, 2H), 2.90 (q, 2H), 2.10 (s, 3H), 1.41 (s, 2H), 1.35-1.22 (m, 4H), 1.18-0.99 (m, 6H), 0.87 (bs, 6H). MS (ESI) m/e 771 (M+H)+. 1.14. Synthesis of 6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4- tetrahydroquinolin-7-yl]-3-{1-[(3-{2-[(2-methoxyethyl)amino]ethoxy}- 5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol-4- yl}pyridine-2-carboxylic acid (Compound W3.14)
1.14.1. 2-((3,5-dimethyl-7-((5-methyl-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-1H-pyrazol-1-yl)methyl)adamantan-1- yl)oxy)ethanol
To a solution of Example 1.1.6 (4.45 g) and PdCl2(dppf)-CH2Cl2 adduct (409 mg) in acetonitrile (60 mL) was added triethylamine (5 mL) and pinacolborane (6.4 mL). The mixture was refluxed overnight. The mixture was used directly in the next step without work up. MS (ESI) m/e 444.80 (M+H)+.
1.14.2. tert-butyl 6-chloro-3-(1-((3-(2-hydroxyethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)picolinate
To a solution of tert-butyl 3-bromo-6-chloropicolinate (3.06 g) in tetrahydrofuran (50 mL) and water (20 mL) was added Example 1.14.1 (4.45 g), 1,3,5,7-tetramethyl-8-tetradecyl-2,4,6-trioxa- 8-phosphaadamantane (0.732 g), Pd2(dba)3 (0.479 g), and K3PO4 (11 g). The mixture was stirred at reflux overnight and concentrated. The residue was dissolved in ethyl acetate (500 mL) and washed with water and brine. The organic layer was dried over Na2SO4, filtered, and concentrated. The residue was purified by flash chromatography, eluting with a gradient of 20-40% ethyl acetate in dichloromethane, to provide the title compound. MS (ESI) m/e 530.23 (M+H)+.
1.14.3. tert-butyl 6-chloro-3-(1-((3,5-dimethyl-7-(2- ((methylsulfonyl)oxy)ethoxy)adamantan-1-yl)methyl)-5-methyl- 1H-pyrazol-4-yl)picolinate
To a cooled (0 °C) stirring solution of Example 1.14.2 (3.88 g) in dichloromethane (30 mL) and triethylamine (6 mL) was added methanesulfonyl chloride (2.52 g). The mixture was stirred at room temperature for 4 hours, diluted with ethyl acetate (400 mL), and washed with water and brine. The organic layer was dried over Na2SO4. Filtration and evaporation of the solvents afforded the title compound. MS (ESI) m/e 608.20 (M+H)+.
1.14.4. tert-butyl 3-(1-((3-(2-aminoethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-chloropicolinate
A solution of Example 1.14.3 (2.2 g) in 7N ammonium in CH3OH (20 mL) was heated at 100 oC under microwave conditions (Biotage Initiator) for 45 minutes and concentrated to dryness. The residue was dissolved in ethyl acetate and washed with water and brine. The organic layer was dried over Na2SO4, filtered, and concentrated to provide the title compound. MS (ESI) m/e 529.33 (M+H)+. 1.14.5. tert-butyl 6-chloro-3-(1-((3,5-dimethyl-7-(2-(2- (trimethylsilyl)ethylsulfonamido)ethoxy)adamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate
To a cooled (0 oC) solution of Example 1.14.4 (3.0 g) in dichloromethane (30 mL) was added triethylamine (3 mL), followed by 2-(trimethylsilyl)ethanesulfonyl chloride (2.3 g). The mixture was stirred at room temperature for 3 hours and concentrated to dryness. The residue was dissolved in ethyl acetate (400 mL) and washed with aqueous NaHCO3, water, and brine. The residue was dried over Na2SO4, filtered, concentrated, and purified by flash chromatography, eluting with 20% ethyl acetate in heptane, to provide the title compound. MS (ESI) m/e 693.04 (M+H)+.
1.14.6. tert-butyl 6-chloro-3-(1-((3-(2-(N-(2-methoxyethyl)-2- (trimethylsilyl)ethylsulfonamido)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)picolinate
To a solution of Example 1.14.5 (415 mg) in toluene (15 mL) was added 2-methoxyethanol (91 mg), followed by cyanomethylenetributylphosphorane (289 mg). The mixture was stirred at 70 oC for 3 hours and concentrated to dryness. The residue was purified by flash chromatography, eluting with 20% ethyl acetate in heptane, to provide the title compound. MS (ESI) m/e 751.04 (M+H)+.
1.14.7. tert-butyl 3-(1-((3-(2-(N-(2-methoxyethyl)-2- (trimethylsilyl)ethylsulfonamido)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (1,2,3,4-tetrahydroquinolin-7-yl)picolinate
To a solution of 7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,2,3,4-tetrahydroquinoline (172 mg) in dioxane (10 mL) and water (5 mL) was added Example 1.14.6 (500 mg), (Ph3P)2PdCl2 (45.6 mg) and CsF (296 mg). The mixture was stirred at 120 oC for 30 minutes under microwave conditions (Biotage Initiator), diluted with ethyl acetate (200 mL) and washed with water and brine. The organic layer was dried over Na2SO4, filtered, and concentrated. The residue was purified by flash chromatography, eluting with 20% ethyl acetate in dichloromethane, to provide the title compound. MS (ESI) m/e 848.09 (M+H)+.
1.14.8. tert-butyl 6-(1-(benzo[d]thiazol-2-ylcarbamoyl)-1,2,3,4- tetrahydroquinolin-7-yl)-3-(1-((3-(2-(N-(2-methoxyethyl)-2- (trimethylsilyl)ethylsulfonamido)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)picolinate
To a suspension of bis(2,5-dioxopyrrolidin-1-yl) carbonate (63 mg) in acetonitrile (10 mL) was added benzo[d]thiazol-2-amine (37.2 mg). The mixture was stirred for 1 hour. A solution of Example 1.14.7 (210 mg) in acetonitrile (2 mL) was added, and the suspension was vigorously stirred overnight diluted with ethyl acetate and washed with water and brine The organic layer was dried over Na2SO4, filtered, and concentrated to provide the title compound. MS (ESI) m/e 1024.50 (M+H)+.
1.14.9. 6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4- tetrahydroquinolin-7-yl]-3-{1-[(3-{2-[(2- methoxyethyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec- 1-yl)methyl]-5-methyl-1H-pyrazol-4-yl}pyridine-2-carboxylic acid
To a solution of Example 1.14.8 (230 mg) in tetrahydrofuran (10 mL) was added tetrabutyl ammonium fluoride (TBAF 10 mL, 1M in tetrahydrofuran). The mixture was stirred at room temperature overnight, diluted with ethyl acetate, and washed with water and brine. The organic layer was dried over Na2SO4, filtered, and concentrated. The residue was dissolved in dichloromethane (5 mL) and treated with trifluoroacetic acid (5 mL) overnight. The mixture was concentrated, and the residue was purified by reverse HPLC (Gilson), eluting with 10-85% acetonitrile in 0.1% TFA/water to provide the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 8.40 (d, 3H), 8.00 (d, 1H), 7.90-7.72 (m, 3H), 7.46 (s, 1H), 7.40-7.32 (m, 1H), 7.28 (d, 1H), 7.24-7.17 (m, 1H), 3.95 (d, 3H), 3.88 (s, 16H), 3.56 (dt, 5H), 3.28 (s, 3H), 3.18-2.96 (m, 5H), 2.82 (t, 2H), 2.21 (s, 3H), 1.93 (p, 2H), 1.43 (s, 2H), 1.30 (q, 5H), 1.21-0.97 (m, 7H), 0.86 (s, 6H) MS (ESI) m/e 804.3 (M+H)+.
1.15. Synthesis of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-3-{1- [(3-{2-[(2-methoxyethyl)amino]ethoxy}-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol-4- yl}pyridine-2-carboxylic acid (Compound W3.15)
1.15.1. 7-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-(N-(2-methoxyethyl)-2- (trimethylsilyl)ethylsulfonamido)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)pyridin-2-yl)-1-naphthoic acid
To a solution of methyl 7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-naphthoate (208 mg) in dioxane (10 mL) and water (5 mL) was added Example 1.14.6 (500 mg), (Ph3P)2PdCl2 (45.6 mg ) and CsF (296 mg). The mixture was stirred at 120 oC for 30 minutes under microwave conditions (Biotage Initiator), diluted with ethyl acetate and washed with water and brine. The organic layer was dried over Na2SO4, filtered, and concentrated. The residue was purified by flash chromatography, eluting with 20% ethyl acetate in dichloromethane, to give the ester intermediate. The ester was dissolved in a mixture of tetrahydrofuran (10 mL), methanol (5 mL) and H2O (5 mL) and treated with lithium hydroxide monohydrate (200 mg). The mixture was stirred at room temperature for 4 hours, acidified with 1N aqueous HCl solution and diluted with ethyl acetate (300 mL). After washing with water ad brine, the organic layer was dried over Na2SO4. After filtration, evaporation of the solvent afforded the title compound. MS (ESI) m/e 888.20 (M+H)+. 1.15.2. 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-3-{1-[(3- {2-[(2-methoxyethyl)amino]ethoxy}-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol- 4-yl}pyridine-2-carboxylic acid
To a solution of Example 1.15.1 (500 mg) in dichloromethane (10 mL) was added benzo[d]thiazol-2-amine (85 mg), 1-ethyl-3-[3-(dimethylamino)propyl]-carbodiimide hydrochloride (216 mg) and 4-(dimethylamino)pyridine (138 mg). The mixture was stirred at room temperature overnight, diluted with ethyl acetate, and washed with water and brine. The organic layer was then dried over Na2SO4, filtered, and concentrated to dryness. The residue was dissolved in
tetrahydrofuran (10 mL) and treated with tetrabutyl ammonium fluoride (10 mL, 1M in
tetrahydrofuran) overnight. The reaction mixture was diluted with ethyl acetate and washed with water and brine. The organic layer was dried over Na2SO4, filtered, and concentrated to dryness. The residue was dissolved in dichloromethane (5 mL) and treated with trifluoroacetic acid (5 mL) overnight. The mixture was then concentrated and the residue was purified by reverse HPLC (Gilson), eluting with 10-85% acetonitrile in 0.1% TFA in water, to give the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 13.11 (s, 1H), 9.00 (s, 1H), 8.60-8.29 (m, 3H), 8.26- 8.13 (m, 3H), 8.03 (ddd, 2H), 7.92 (d, 1H), 7.80 (d, 1H), 7.74-7.62 (m, 1H), 7.51-7.42 (m, 2H), 7.36 (td, 1H), 3.88 (s, 2H), 3.61-3.52 (m, 2H), 3.27 (s, 3H), 3.17-2.95 (m, 4H), 2.22 (s, 3H), 1.43 (s, 2H), 1.30 (q, 4H), 1.23-0.96 (m, 6H), 0.86 (s, 6H). MS (ESI) m/e 799.2 (M+H)+.
1.16. Synthesis of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]-3-[1-({3,5-dimethyl-7-[2-(oxetan-3- ylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol- 4-yl]pyridine-2-carboxylic acid (Compound W3.16)
1.16.1. methyl 2-(5-bromo-6-(tert-butoxycarbonyl)pyridin-2-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxylate
To a solution of methyl 1,2,3,4-tetrahydroisoquinoline-8-carboxylate hydrochloride (12.37 g) and Example 1.4.4 (15 g) in dimethyl sulfoxide (100 mL) was added N,N-diisopropylethylamine(12 mL). The mixture was stirred at 50 oC for 24 hours. The mixture was diluted with ethyl acetate (500 mL), washed with water and brine, and dried over Na2SO4. After filtration and evaporation of the solvent, the crude material was purified via silica gel column chromatography, eluting with 20% ethyl acetate in hexane, to give the title compound. MS (ESI) m/e 448.4 (M+H)+.
1.16.2. methyl 2-(6-(tert-butoxycarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyridin-2-yl)-1,2,3,4-tetrahydroisoquinoline-8- carboxylate
To a solution of Example 1.16.1 (2.25 g) and [1,1'- bis(diphenylphosphino)ferrocene]dichloropalladium(II) (205 mg) in acetonitrile (30 mL) was added triethylamine (3 mL) and pinacolborane (2 mL) The mixture was stirred at reflux for 3 hours The mixture was diluted with ethyl acetate (200 mL), washed with water and brine, and dried over Na2SO4. Filtration, evaporation of the solvent, and silica gel chromatography (eluting with 20% ethyl acetate in hexane) gave the title compound. MS (ESI) m/e 495.4 (M+H)+.
1.16.3. methyl 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxylate
To a solution of Example 1.16.2 (4.94 g) in tetrahydrofuran (60 mL) and water (20 mL) was added Example 1.4.2 (5.57 g), 1,3,5,7-tetramethyl-8-tetradecyl-2,4,6-trioxa-8-phosphaadamantane (412 mg), tris(dibenzylideneacetone)dipalladium(0) (457 mg), and K3PO4 (11 g). The mixture was stirred at reflux overnight. The reaction mixture was diluted with ethyl acetate (500 mL), washed with water and brine, and dried over Na2SO4. After filtration and evaporation of the solvent, the crude material was purified via column chromatography, eluting with 20% ethyl acetate in heptane, to give the title compound. MS (ESI) m/e 784.4 (M+H)+.
1.16.4. 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxylic acid
To a solution of Example 1.16.3 (10 g) in tetrahydrofuran (60 mL), methanol (30 mL) and water (30 mL), was added lithium hydroxide monohydrate (1.2 g). The mixture was stirred at room temperature for 24 hours. The reaction mixture was neutralized with 2% aqueous HCl and concentrated under vacuum. The residue was diluted with ethyl acetate (800 mL), washed with water and brine, and dried over Na2SO4. Filtration and evaporation of the solvent gave the title compound. MS (ESI) m/e 770.4 (M+H)+.
1.16.5. tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-((tert- butoxycarbonyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate
To a solution of Example 1.16.4 (3.69 g) in N,N-dimethylformamide (20 mL) was added benzo[d]thiazol-2-amine(1.1 g), fluoro-N,N,N',N'-tetramethylformamidinium hexafluorophosphate (1.9 g) and N,N diisopropylethylamine (1.86 g). The mixture was stirred at 60 °C for 3 hours. The reaction mixture was diluted with ethyl acetate (500 mL), washed with water and brine, and dried over Na2SO4. Filtration, evaporation of the solvent, and column purification (20% ethyl acetate in heptane) gave the title compound. MS (ESI) m/e 902.2(M+H)+.
1.16.6. 3-(1-((3-(2-aminoethoxy)-5,7-dimethyladamantan-1-yl)methyl)-5- methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 34-dihydroisoquinolin-2(1H)-yl)picolinic acid Example 1.16.5 (2 g) was dissolved in 50% TFA in dichloromethane (20 mL) and stirred overnight. The solvents were removed under vacuum and the residue was loaded on a reverse-phase column and eluted with 20-80% acetonitrile in water (0.1% TFA) to give the title compound. MS (ESI) m/e 746.3 (M+H)+.
1.16.7. 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl]-3-[1-({3,5-dimethyl-7-[2-(oxetan-3- ylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H- pyrazol-4-yl]pyridine-2-carboxylic acid
A solution of Example 1.16.6 (0.050 g), oxetan-3-one (5 mg) and sodium
triacetoxyborohydride (0.018 g) was stirred together in dichloromethane (1 mL) at room temperature. After stirring for 1 hour, additional oxetan-3-one (5 mg) and sodium triacetoxyborohydride (0.018 g) were added and the reaction was stirred overnight. The reaction was concentrated, dissolved in a 1:1 mixture of dimethyl sulfoxide/methanol (2 mL) and purified by HPLC using a Gilson system (20-60% acetonitrile in water containing 0.1% v/v trifluoroacetic acid). The desired fractions were combined and freeze-dried to provide the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 12.95 (s, 1H), 9.26 (s, 2H), 8.12 (d, 1H), 7.88 (d, 1H), 7.71 (d, 1H), 7.63-7.50 (m, 3H), 7.50-7.41 (m, 2H), 7.38 (s, 1H), 7.05 (d, 1H), 5.05 (s, 2H), 4.79 (t, 2H), 4.68 (dd, 2H), 4.54-4.41 (m, 1H), 3.98 (t, 2H), 3.92 (s, 2H), 3.63 (t, 2H), 3.16-3.04 (m, 4H), 2.20 (s, 3H), 1.52 (s, 2H), 1.47-1.06 (m, 10H), 0.96 (s, 6H). MS (ESI) m/e 802.2 (M+H)+.
1.17. Synthesis of 6-[6-(3-aminopyrrolidin-1-yl)-8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-(1-{[3-(2- methoxyethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5- methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid (Compound W3.17) 1.17.1. 4-iodo-1-((3-(2-methoxyethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazole
Example 1.1.6 (3.00 g) was dissolved in 1,4-dioxane (40 mL), and sodium hydride (60% in mineral oil, 568 mg) was added. The solution was mixed at room temperature for 15 minutes, and methyl iodide (1.64 mL) was added. The solution was stirred at room temperature for three days, and then 0.01 M aqueous HCl solution (50 mL) was added. The solution was extracted with diethyl ether three times. The combined organic extracts were washed with brine and dried on anhydrous sodium sulfate. After filtration, the solvent was removed under reduced pressure and then under high vacuum to yield the title compound. MS (ESI) m/e 459 (M+H)+.
1.17.2. benzyl 4-oxopent-2-ynoate
Benzyl 4-hydroxypent-2-ynoate (40.5 g) and Dess-Martin Periodinane (93.0 g) in dichloromethane (500 mL) were stirred for 1 hour at 0 °C. The solution was poured into diethyl ether (1L), and the combined organics were washed three times with 1M aqueous NaOH and brine, dried over Na2SO4, filtered, and concentrated. The residue was chromatographed on silica gel using 5% ethyl acetate in heptanes to give the title compound.
1.17.3. (S)-benzyl 6-(3-((tert-butoxycarbonyl)amino)pyrrolidin-1-yl)-2- (2,2,2-trifluoroacetyl)-1,2,3,4-tetrahydroisoquinoline-8- carboxylate
A solution of 1-(2,2,2-trifluoroacetyl)piperidin-4-one (6.29 g), (S)-tert-butyl pyrrolidin-3- ylcarbamate (6.0 g), and p-toluenesulfonic acid monohydrate (0.613 g) in ethanol (80 mL) was stirred for 1 hour at room temperature. Example 1.17.2 (6.51 g) was then added and the reaction was stirred for 24 hours at room temperature, and heated to 45 oC for 3 days. The reaction was then cooled and poured into diethyl ether (600 mL). The resulting solution was washed twice with water and brine, dried over Na2SO4, filtered, and concentrated. The residue was chromatographed on silica gel using 5-50% ethyl acetate in heptanes to give the product.
1.17.4. (S)-benzyl 6-(3-((tert-butoxycarbonyl)amino)pyrrolidin-1-yl)- 1,2,3,4-tetrahydroisoquinoline-8-carboxylate
A solution of Example 1.17.3 (3.1 g) and potassium carbonate (1.8 g) in a mixture of tetrahydrofuran (30 mL), methanol (10 mL), and water (25 mL) was stirred for 48 hours at 45 °C. The reaction was then cooled and diluted with dichloromethane (300 mL). The layers were separated and the organic layer was dried over Na2SO4, filtered, and concentrated to give the title compound.
1.17.5. (S)-benzyl 2-(5-bromo-6-(tert-butoxycarbonyl)pyridin-2-yl)-6-(3- ((tert-butoxycarbonyl)amino)pyrrolidin-1-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxylate
A solution of Example 1.17.4 (1.6 g), Example 1.4.4 (1.08 g), and triethylamine (0.59 mL) in N,N-dimethylformamide (10 mL) was heated to 50 oC for 24 hours. The reaction was cooled and poured into ethyl acetate (400 mL). The resulting solution was washed three times with water and brine, dried over Na2SO4, filtered, and concentrated. The residue was chromatographed on silica gel using 5-50% ethyl acetate in heptanes to give the product.
1.17.6. (S)-benzyl 2-(6-(tert-butoxycarbonyl)-5-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)pyridin-2-yl)-6-(3-((tert- butoxycarbonyl)amino)pyrrolidin-1-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxylate
A solution of Example 1.17.5 (500 mg), 4,4,5,5-tetramethyl-1,3,2-dioxaborolane (136 mg), and triethylamine (0.200 mL) in acetonitrile (5 mL) was heated to 75 oC for 24 hours. The reaction was allowed to cool to room temperature and concentrated to dryness. The crude material was then purified via column chromatography, eluting with 5-50% ethyl acetate in heptanes, to give the title compound.
1.17.7. benzyl 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-methoxyethoxy)- 57-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)pyridin-2-yl)-6-((S)-3-((tert-butoxycarbonyl)amino)pyrrolidin- 1-yl)-1,2,3,4-tetrahydroisoquinoline-8-carboxylate
A solution of Example 1.17.6 (240 mg), Example 1.17.1 (146 mg), 1,3,5,7-tetramethyl-8- tetradecyl-2,4,6-trioxa-8-phosphaadamantane (13 mg), palladium (II)acetate (14.6 mg), and tripotassium phosphate (270 mg) in dioxane (7 mL) and water (3 mL) was heated to 70 oC for 24 hours. The reaction was allowed to cool to room temperature and was concentrated to dryness. The crude material was then purified via column chromatography, eluting with 5-25% ethyl acetate in heptanes, to give the title compound.
1.17.8. 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-methoxyethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)pyridin-2-yl)-6-((S)-3-((tert-butoxycarbonyl)amino)pyrrolidin- 1-yl)-1,2,3,4-tetrahydroisoquinoline-8-carboxylic acid
A solution of Example 1.17.7 (1.6 g) and lithium hydroxide monohydrate (5 mg) in a 3:1:1 mixture of tetrahydrofuran/methanol/water (10 mL) was stirred for 4 days. The reaction was acidified with 1M aqueous HCl solution and poured into ethyl acetate (150 mL). The resulting solution was washed with brine, dried over Na2SO4, filtered, and concentrated to give the title compound.
1.17.9. tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-6-((S)-3-((tert- butoxycarbonyl)amino)pyrrolidin-1-yl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(1-((3-(2-methoxyethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate
A solution of Example 1.17.8 (78 mg), benzo[d]thiazol-2-amine (16 mg), O-(7- azabenzotriazol-1-yl)-N,N,N’,N’-tetramethyluronium hexafluorophosphate (48 mg), and
diisopropylethylamine (0.024 mL) in N,N-dimethylformamide (3 mL) was heated to 50 oC for 48 hours. The reaction was then cooled and poured into ethyl acetate (100 mL). The resulting solution was washed three times with water and brine, dried over Na2SO4, filtered, and concentrated. The residue was purified via column chromatography, eluting with 20-100% ethyl acetate in heptanes, to give the title compound.
1.17.10. 6-[6-(3-aminopyrrolidin-1-yl)-8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-(1-{[3-(2- methoxyethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}- 5-methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid
Example 1.17.9 (40 mg) in dichloromethane (3 mL) was treated with trifluoroacetic acid (2 mL) overnight. The mixture was concentrated to provide the title compound as a TFA salt. MS (ESI) m/e 845.7 (M+H)+.
1.18. Synthesis of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]-3-{1-[(3,5-dimethyl-7-{2-[(2- sulfamoylethyl)amino]ethoxy}tricyclo[3.3.1.13,7]dec-1-yl)methyl]-5- methyl-1H-pyrazol-4-yl}pyridine-2-carboxylic acid (Compound W3.18) 1.18.1. 3-bromo-5,7-dimethyladamantanecarboxylic acid
Into a 50 mL round-bottomed flask at 0 °C, was added bromine (16 mL). Iron powder (7 g) was added, and the reaction was stirred at 0 °C for 30 minutes. 3,5-Dimethyladamantane-1- carboxylic acid (12 g) was added. The mixture was warmed up to room temperature and stirred for 3 days. A mixture of ice and concentrated HCl was poured into the reaction mixture. The resulting suspension was treated twice with Na2SO3 (50 g in 200 mL water) and extracted three times with dichloromethane. The combined organics were washed with 1N aqueous HCl, dried over sodium sulfate, filtered, and concentrated to give the title compound.
1.18.2. 3-bromo-5,7-dimethyladamantanemethanol
To a solution of Example 1.18.1 (15.4 g) in tetrahydrofuran (200 mL) was added BH3 (1M in tetrahydrofuran, 150 mL), and the mixture was stirred at room temperature overnight. The reaction mixture was then carefully quenched by adding methanol dropwise. The mixture was then concentrated under vacuum, and the residue was balanced between ethyl acetate (500 mL) and 2N aqueous HCl (100 mL). The aqueous layer was further extracted twice with ethyl acetate, and the combined organic extracts were washed with water and brine, dried over sodium sulfate, and filtered. Evaporation of the solvent gave the title compound.
1.18.3. 1-((3-bromo-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl)-1H- pyrazole
To a solution of Example 1.18.2 (8.0 g) in toluene (60 mL) was added 1H-pyrazole (1.55 g) and cyanomethylenetributylphosphorane (2.0 g), and the mixture was stirred at 90 oC overnight. The reaction mixture was concentrated, and the residue was purified by silica gel column chromatography (10:1 heptane:ethyl acetate) to give the title compound. MS (ESI) m/e 324.2 (M+H)+.
1.18.4. 2-{[3,5-dimethyl-7-(1H-pyrazol-1-ylmethyl)tricyclo[3.3.1.13,7]dec- 1-yl]oxy}ethanol
To a solution of Example 1.18.3 (4.0 g) in ethane-1,2-diol (12 mL) was added triethylamine (3 mL). The mixture was stirred at 150 oC under microwave conditions (Biotage Initiator) for 45 minutes. The mixture was poured into water (100 mL) and extracted three times with ethyl acetate. The combined organic extracts were washed with water and brine, dried over sodium sulfate, and filtered. Evaporation of the solvent gave a residue that was purified by silica gel chromatography, eluting with 20% ethyl acetate in heptane, followed by 5% methanol in dichloromethane, to give the title compound. MS (ESI) m/e 305.2 (M+H)+.
1.18.5. 2-({3,5-dimethyl-7-[(5-methyl-1H-pyrazol-1- yl)methyl]tricyclo[3.3.1.13,7]dec-1-yl}oxy)ethanol
To a cooled (-78 oC) solution of Example 1.18.4 (6.05 g) in tetrahydrofuran (100 mL) was added n-BuLi (40 mL 25M in hexane) and the mixture was stirred at-78 oC for 15 hours Iodomethane (10 mL) was added through a syringe, and the mixture was stirred at-78 oC for 3 hours. The reaction mixture was then quenched with aqueous NH4Cl and extracted twice with ethyl acetate, and the combined organic extracts were washed with water and brine. After drying over sodium sulfate, the solution was filtered and concentrated, and the residue was purified by silica gel column chromatography, eluting with 5% methanol in dichloromethane, to give the title compound. MS (ESI) m/e 319.5 (M+H)+.
1.18.6. 1-({3,5-dimethyl-7-[2-(hydroxy)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-4-iodo-5-methyl-1H-pyrazole
To a solution of Example 1.18.5 (3.5 g) in N,N-dimethylformamide (30 mL) was added N- iodosuccinimide (3.2 g), and the mixture was stirred at room temperature for 1.5 hours. The reaction mixture was diluted with ethyl acetate (600 mL) and washed with aqueous NaHSO3, water and brine. The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with 20% ethyl acetate in
dichloromethane, to give the title compound. MS (ESI) m/e 445.3 (M+H)+.
1.18.7. 1-((3-(2-((tert-butyldimethylsilyl)oxy)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-4-iodo-5-methyl-1H-pyrazole
Tert-butyldimethylsilyl trifluoromethanesulfonate (5.34 mL) was added to a solution of Example 1.18.6 (8.6 g) and 2,6-lutidine (3.16 mL) in dichloromethane (125 mL) at-40 oC, and the reaction was allowed to warm to room temperature overnight. The mixture was concentrated, and the residue was purified by silica gel chromatography, eluting with 5-20% ethyl acetate in heptanes, to give the title compound. MS (ESI) m/e 523.4 (M+H)+.
1.18.8. 1-((3-(2-((tert-butyldimethylsilyl)oxy)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole
n-Butyllithium (8.42 mL, 2.5M in hexanes) was added to Example 1.18.7 (9.8 g) in 120 mL tetrahydrofuran at-78 °C, and the reaction was stirred for 1 minute. Trimethyl borate (3.92 mL) was added, and the reaction stirred for 5 minutes. Pinacol (6.22 g) was added, and the reaction was allowed to warm to room temperature and was stirred 2 hours. The reaction was quenched with pH 7 buffer, and the mixture was poured into ether. The layers were separated, and the organic layer was concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with 1-25% ethyl acetate in heptanes, to give the title compound.
1.18.9. 6-fluoro-3-bromopicolinic acid
A slurry of 6-amino-3-bromopicolinic acid (25 g) in 400 mL 1:1 dichloromethane/chloroform was added to nitrosonium tetrafluoroborate (18.2 g) in dichloromethane (100 mL) at 5 oC over 1 hour. The resulting mixture was stirred for another 30 minutes, then warmed to 35 oC and stirred overnight. The reaction was cooled to room temperature, and then adjusted to pH 4 with aqueous NaH2PO4 solution The resulting solution was extracted three times with dichloromethane and the combined extracts were washed with brine, dried over sodium sulfate, filtered and concentrated to provide the title compound.
1.18.10. Tert-butyl 3-bromo-6-fluoropicolinate
Para-toluenesulfonyl chloride (27.6 g) was added to a solution of Example 1.18.9 (14.5 g) and pyridine (26.7 mL) in dichloromethane (100 mL) and tert-butanol (80 mL) at 0 °C. The reaction was stirred for 15 minutes, and then warmed to room temperature, and stirred overnight. The solution was concentrated and partitioned between ethyl acetate and aqueous Na2CO3 solution. The layers were separated, and the aqueous layer extracted with ethyl acetate. The organic layers were combined, rinsed with aqueous Na2CO3 solution and brine, dried over sodium sulfate, filtered, and concentrated to provide the title compound.
1.18.11. methyl 2-(5-bromo-6-(tert-butoxycarbonyl)pyridin-2-yl)- 1,2,3,4-tetrahydroisoquinoline-8-carboxylate
To a solution of methyl 1,2,3,4-tetrahydroisoquinoline-8-carboxylate hydrochloride (12.37 g) and Example 1.18.10 (15 g) in dimethyl sulfoxide (100 mL) was added N,N-diisopropylethylamine (12 mL), and the mixture was stirred at 50 oC for 24 hours. The mixture was then diluted with ethyl acetate (500 mL) and washed with water and brine. The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel
chromatography, eluting with 20% ethyl acetate in hexane, to give the title compound. MS (ESI) m/e 448.4 (M+H)+.
1.18.12. methyl 2-(6-(tert-butoxycarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyridin-2-yl)-1,2,3,4-tetrahydroisoquinoline- 8-carboxylate
To a solution of Example 1.18.11 (2.25 g) and [1,1'- bis(diphenylphosphino)ferrocene]dichloropalladium(II)(205 mg) in acetonitrile (30 mL) was added triethylamine (3 mL) and pinacolborane (2 mL), and the mixture was stirred at reflux for 3 hours. The mixture was diluted with ethyl acetate (200 mL) and washed with water and brine. The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure. Purification of the residue by silica gel chromatography, eluting with 20% ethyl acetate in hexane, provided the title compound.
1.18.13. methyl 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-hydroxyethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)pyridin-2-yl)-1,2,3,4-tetrahydroisoquinoline-8-carboxylate
To a solution of Example 1.18.12 (2.25 g) in tetrahydrofuran (30 mL) and water (10 mL) was added Example 1.18.6 (2.0 g), 1,3,5,7-tetramethyl-6-phenyl-2,4,8-trioxa-6-phosphaadamantane (329 mg), tris(dibenzylideneacetone)dipalladium(0) (206 mg) and potassium phosphate tribasic (4.78 g). The mixture was refluxed overnight, cooled and diluted with ethyl acetate (500 mL). The resulting mixture was washed with water and brine and the organic layer was dried over sodium sulfate filtered and concentrated. The residue was purified by flash chromatography, eluting with 20% ethyl acetate in heptanes followed by 5% methanol in dichloromethane, to provide the title compound.
1.18.14. methyl 2-(6-(tert-butoxycarbonyl)-5-(1-((3,5-dimethyl-7-(2- ((methylsulfonyl)oxy)ethoxy)adamantan-1-yl)methyl)-5- methyl-1H-pyrazol-4-yl)pyridin-2-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxylate
To a cold solution of Example 1.18.13 (3.32 g) in dichloromethane (100 mL) in an ice-bath was sequentially added triethylamine (3 mL) and methanesulfonyl chloride (1.1 g). The reaction mixture was stirred at room temperature for 1.5 hours and diluted with ethyl acetate, and washed with water and brine. The organic layer was dried over sodium sulfate, filtered, and concentrated to provide the title compound.
1.18.15. methyl 2-(5-(1-((3-(2-azidoethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(tert- butoxycarbonyl)pyridin-2-yl)-1,2,3,4-tetrahydroisoquinoline-8- carboxylate
To a solution of Example 1.18.14 (16.5 g) in N,N-dimethylformamide (120 mL) was added sodium azide (4.22 g). The mixture was heated at 80 oC for 3 hours, cooled, diluted with ethyl acetate and washed with water and brine. The organic layer was dried over sodium sulfate, filtered, and concentrated. The residue was purified by flash chromatography, eluting with 20% ethyl acetate in heptanes, to provide the title compound.
1.18.16. 2-(5-(1-((3-(2-azidoethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(tert- butoxycarbonyl)pyridin-2-yl)-1,2,3,4-tetrahydroisoquinoline-8- carboxylic acid
To a solution of Example 1.18.15 (10 g) in a mixture of tetrahydrofuran (60 mL), methanol (30 mL) and water (30 mL) was added lithium hydroxide monohydrate (1.2g). The mixture was stirred at room temperature overnight and neutralized with 2% aqueous HCl. The resulting mixture was concentrated, and the residue was dissolved in ethyl acetate (800 mL), and washed with brine. The organic layer was dried over sodium sulfate, filtered, and concentrated to provide the title compound.
1.18.17. tert-butyl 3-(1-((3-(2-azidoethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)picolinate
A mixture of Example 1.18.16 (10 g), benzo[d]thiazol-2-amine (3.24 g), fluoro-N,N,N',N'- tetramethylformamidinium hexafluorophosphate (5.69 g) and N,N-diisopropylethylamine (5.57 g) in N,N-dimethylformamide (20 mL) was heated at 60 °C for 3 hours, cooled and diluted with ethyl acetate The resulting mixture was washed with water and brine The organic layer was dried over sodium sulfate, filtered, and concentrated. The residue was purified by flash chromatography, eluting with 20% ethyl acetate in dichloromethane to give the title compound.
1.18.18. tert-butyl 3-(1-(((3-(2-aminoethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)picolinate
To a solution of Example 1.18.17 (2.0 g) in tetrahydrofuran (30 mL) was added Pd/C (10%, 200 mg). The mixture was stirred under a hydrogen atmosphere overnight. The insoluble material was filtered off and the filtrate was concentrated to provide the title compound.
1.18.19. 3-(1-((3-(2-aminoethoxy)-5,7-dimethyladamantan-1-yl)methyl)- 5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)picolinic acid
Example 1.18.18 (200 mg) in dichloromethane (2.5 mL) was treated with trifluoroacetic acid (2.5 mL) overnight. The reaction mixture was concentrated, and the residue was purified by reverse phase chromatography (C18 column), eluting with 20-60% acetonitrile in water containing 0.1% v/v trifluoroacetic acid, to provide the title compound. MS (ESI) m/e 746.2 (M+H)+.
1.18.20. 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl]-3-{1-[(3,5-dimethyl-7-{2-[(2- sulfamoylethyl)amino]ethoxy}tricyclo[3.3.1.13,7]dec-1- yl)methyl]-5-methyl-1H-pyrazol-4-yl}pyridine-2-carboxylic acid
A mixture of Example 1.18.19 (18 mg) and ethenesulfonamide (5.2 mg) in N,N- dimethylformamide (1 mL) and water (0.3 mL) was stirred for one week. The mixture was purified by reverse phase chromatography (C18 column), eluting with 20-60% acetonitrile in water containing 0.1% v/v trifluoroacetic acid, to provide the title compound. 1H NMR (500 MHz, dimethyl sulfoxide- d6) δ ppm 8.03 (d, 1H), 7.79 (d, 1H), 7.61 (d, 1H), 7.45-7.50 (m, 1H), 7.41-7.44 (m, 1H), 7.33-7.39 (m, 3H), 7.23 (s, 1H), 6.73 (d, 1H), 4.87 (s, 2H), 3.89 (t, 2H), 3.79 (s, 2H), 3.12-3.20 (m, 2H), 2.99 (t, 2H), 2.85 (s, 2H), 2.09 (s, 3H), 1.32 (dd, 4H), 1.08-1.19 (m, 5H), 1.04 (d, 4H), 0.86 (s, 6H). MS (ESI) m/e 853.2(M+H)+.
1.19 Synthesis of 3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[3-(1,3-benzothiazol-2-ylcarbamoyl)-6,7- dihydrothieno[3,2-c]pyridin-5(4H)-yl]pyridine-2-carboxylic acid (W3.19) 1.19.1 6,7-dihydro-4H-thieno[3,2-c]pyridine-3,5-dicarboxylic acid 5-tert-butyl ester 3-methyl ester
Tert-butyl 3-bromo-6,7-dihydrothieno[3,2-c]pyridine-5(4H)-carboxylate (1000 mg) and dichloro[1,1’-bis(diphenylphosphino)ferrocene]palladium(II) (69 mg) were placed in a 50 mL pressure bottle, and methanol (20 mL) was added, followed by trimethylamine (636 mg). The solution was degassed and flushed with argon three times. The solution was then degassed and flushed with carbon monoxide and heated to 100 °C for 18 hours under 60 psi of carbon monoxide The solvent was removed under reduced pressure, and the residue was purified by flash column chromatography on silica gel, eluting with 50% ethyl acetate in heptanes. The solvent was removed under reduced pressure to yield the title compound.
1.19.2 4,5,6,7-tetrahydro-thieno[3,2-c]pyridine-3-carboxylic acid methyl ester Example 1.19.1 (940 mg) was dissolved in dichloromethane (12 mL). Trifluoroacetic acid (2220 mg) was added, and the solution was stirred for three hours. The solvent was removed under reduced pressure to yield the title compound as the trifluoroacetic acid salt, which was used without further purification.
1.19.3 5-(5-bromo-6-tert-butoxycarbonyl-pyridin-2-yl)-4,5,6,7-tetrahydro- thieno[3,2-c]pyridine-3-carboxylic acid methyl ester
The title compound was prepared by substituting Example 1.19.2 for ethyl 5,6,7,8- tetrahydroimidazo[1,5-a]pyrazine-1-carboxylate hydrochloride in Example 1.4.5. MS (ESI) m/e 452, 450 (M+H)+.
1.19.4 5-[6-tert-butoxycarbonyl-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2- yl)-pyridin-2-yl]-4,5,6,7-tetrahydro-thieno[3,2-c]pyridine-3-carboxylic acid methyl ester
The title compound was prepared by substituting Example 1.19.3 for Example 1.1.9 in Example 1.1.10. MS (ESI) m/e 500 (M+H)+, 531 (M+CH3OH-H)-.
1.19.5 5-(6-tert-butoxycarbonyl-5-{1-[5-(2-tert-butoxycarbonylamino- ethoxy)-3,7-dimethyl-adamantan-1-ylmethyl]-5-methyl-1H-pyrazol-4- yl}-pyridin-2-yl)-4,5,6,7-tetrahydro-thieno[3,2-c]pyridine-3-carboxylic acid methyl ester
The title compound was prepared by substituting Example 1.19.4 for Example 1.4.6 in Example 1.4.7.
1.19.6 5-(6-tert-butoxycarbonyl-5-{1-[5-(2-tert-butoxycarbonylamino- ethoxy)-3,7-dimethyl-adamantan-1-ylmethyl]-5-methyl-1H-pyrazol-4- yl}-pyridin-2-yl)-4,5,6,7-tetrahydro-thieno[3,2-c]pyridine-3-carboxylic acid
The title compound was prepared by substituting Example 1.19.5 for Example 1.4.7 in Example 1.4.8. MS (ESI) m/e 776 (M+H)+, 774 (M-H)-.
1.19.7 6-[3-(benzothiazol-2-ylcarbamoyl)-6,7-dihydro-4H-thieno[3,2- c]pyridin-5-yl]-3-{1-[5-(2-tert-butoxycarbonylamino-ethoxy)-3,7- dimethyl-adamantan-1-ylmethyl]-5-methyl-1H-pyrazol-4-yl}-pyridine- 2-carboxylic acid tert-butyl ester
The title compound was prepared by substituting Example 1.19.6 for Example 1.4.8 in Example 1.4.9. MS (ESI) m/e 892 (M+H)+, 890 (M-H)-. 1.19.8 3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[3-(1,3-benzothiazol-2- ylcarbamoyl)-6,7-dihydrothieno[3,2-c]pyridin-5(4H)-yl]pyridine-2- carboxylic acid
The title compound was prepared by substituting Example 1.19.7 for Example 1.1.13 in Example 1.1.14. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 8.11 (bs, 1H), 8.00 (d, 1H), 7.77 (d, 1H), 7.68 (bs, 3H), 7.53 (d, 1H), 7.47 (t, 1H), 7.36-7.31 (m, 2H), 7.14 (d, 1H), 4.71 (s, 2H), 3.99 (t, 2H), 3.85 (s, 2H), 3.52 (m, 2H), 3.00 (t, 2H), 2.91 (q, 2H), 2.13 (s, 3H), 1.44 (s, 2H), 1.31 (q, 4H), 1.16 (m, 4H), 1.05 (q, 2H), 0.88 (s, 6H). MS (ESI) m/e 752 (M+H)+, 750 (M-H)-.
1.20 Synthesis of 3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-3- (trifluoromethyl)-5,6-dihydroimidazo[1,5-a]pyrazin-7(8H)-yl]pyridine-2- carboxylic acid (W3.20)
1.20.1 7-(5-bromo-6-tert-butoxycarbonyl-pyridin-2-yl)-3-trifluoromethyl- 5,6,7,8-tetrahydro-imidazo[1,5-a]pyrazine-1-carboxylic acid methyl ester
The title compound was prepared by substituting methyl 3-(trifluoromethyl)-5,6,7,8- tetrahydroimidazo[1,5-a]pyrazine-1-carboxylate for ethyl 5,6,7,8-tetrahydroimidazo[1,5-a]pyrazine-1- carboxylate hydrochloride in Example 1.4.5. MS (ESI) m/e 449 (M-tBu+H)+, 503 (M-H)-.
1.20.2 7-[6-tert-butoxycarbonyl-5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2- yl)-pyridin-2-yl]-3-trifluoromethyl-5,6,7,8-tetrahydro-imidazo[1,5- a]pyrazine-1-carboxylic acid methyl ester
The title compound was prepared by substituting Example 1.20.1 for Example 1.1.9 in Example 1.1.10. MS (ESI) m/e 553 (M+H)+.
1.20.3 di-tert-butyl [2-({3-[(4-iodo-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]decan-1-yl}oxy)ethyl]-2-imidodicarbonate Example 1.1.6 (5.000 g) was dissolved in dichloromethane (50 mL). Triethylamine (1.543 g) was added, and the solution was cooled on an ice bath. Methanesulfonyl chloride (1.691 g) was added dropwise. The solution was allowed to warm to room temperature and stir for 30 minutes. Saturated aqueous sodium bicarbonate solution (50 mL) was added. The layers were separated, and the organic layer was washed with brine (50 mL). The aqueous portions were then combined and back extracted with dichloromethane (50 mL). The organic portions were combined, dried over anhydrous sodium sulfate, filtered, and concentrated. The residue was dissolved in acetonitrile (50 mL). Di-tert-butyl iminodicarboxylate (2.689 g) and cesium carbonate (7.332 g) were added, and the solution was refluxed for 16 hours. The solution was cooled and added to diethyl ether (100 mL) and water (100 mL). The layers were separated. The organic portion was washed with brine (50 mL). The aqueous portions were then combined and back extracted with diethyl ether (100 mL) The organic portions were combined, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. The material was purified by flash column chromatography on silica gel, eluting with 20% ethyl acetate in heptanes. The solvent was evaporated under reduced pressure to provide the title compound. MS (ESI) m/e 666 (M+Na)+.
1.20.4 methyl 7-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-(di-(tert- butoxycarbonyl)amino)ethoxy)-5,7-dimethyladamantan-1-yl)methyl)- 5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-3-(trifluoromethyl)-5,6,7,8- tetrahydroimidazo[1,5-a]pyrazine-1-carboxylate
The title compound was prepared by substituting Example 1.20.2 for Example 1.4.6 and Example 1.20.3 for Example 1.4.2 in Example 1.4.7. MS (ESI) m/e 964 (M+Na)+, 940 (M-H)-.
1.20.5 7-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-(di-(tert- butoxycarbonyl)amino)ethoxy)-5,7-dimethyladamantan-1-yl)methyl)- 5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-3-(trifluoromethyl)-5,6,7,8- tetrahydroimidazo[1,5-a]pyrazine-1-carboxylic acid
The title compound was prepared by substituting Example 1.20.4 for Example 1.4.7 in Example 1.4.8. MS (ESI) m/e 828 (M+H)+, 826 (M-H)-.
1.20.6 tert-butyl 6-(1-(benzo[d]thiazol-2-ylcarbamoyl)-3-(trifluoromethyl)- 5,6-dihydroimidazo[1,5-a]pyrazin-7(8H)-yl)-3-(1-((3-(2-(di-(tert- butoxycarbonyl)amino)ethoxy)-5,7-dimethyladamantan-1-yl)methyl)- 5-methyl-1H-pyrazol-4-yl)picolinate
The title compound was prepared by substituting Example 1.20.5 for Example 1.4.8 in Example 1.4.9. MS (ESI) m/e 1058 (M-H)-.
1.20.7 3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[1-(1,3-benzothiazol-2- ylcarbamoyl)-3-(trifluoromethyl)-5,6-dihydroimidazo[1,5-a]pyrazin- 7(8H)-yl]pyridine-2-carboxylic acid
The title compound was prepared by substituting Example 1.20.6 for Example 1.1.13 in Example 1.1.14. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 11.99 (bs, 1H), 8.00 (d, 1H), 7.79 (d, 1H), 7.66 (bs, 3H), 7.61 (d, 1H), 7.47 (t, 1H), 7.35 (t, 2H), 7.19 (d, 1H), 5.20 (s, 2H), 4.37 (t, 2H), 4.16 (t, 2H), 3.86 (s, 2H), 3.51 (t, 2H), 2.91 (q, 2H), 2.14 (s, 3H), 1.44 (s, 2H), 1.36-1.24 (m, 4H), 1.19-1.02 (m, 6H), 0.88 (s, 6H). MS (ESI) m/e 804 (M+H)+, 802 (M-H)-.
1.21 Synthesis of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-6-{methyl[2- (methylamino)ethyl]amino}-3,4-dihydroisoquinolin-2(1H)-yl]-3-(1-{[3-(2- methoxyethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)pyridine-2-carboxylic acid (W3.21)
1.21.1 methyl 3-bromo-5-(bromomethyl)benzoate AIBN (2,2’-azobis(2-methylpropionitrile)) (1.79 g) was added to methyl 3-bromo-5- methylbenzoate (50 g) and N-bromosuccinimide (44.7 g) in 350 mL acetonitrile, and the mixture was refluxed overnight. An additional 11 g of N-bromosuccinimide and 0.5 g of AIBN (2,2’-azobis(2- methylpropionitrile)) was added, and the refluxing was continued for 3 hours. The mixture was concentrated, and then taken up in 500 mL ether, and stirred for 30 minutes. The mixture was then filtered, and the resulting solution was concentrated. The crude product was chromatographed on silica gel using 10% ethyl acetate in heptane to give the title compound.
1.21.2 methyl 3-bromo-5-(cyanomethyl)benzoate
Tetrabutylammonium cyanide (50 g) was added to Example 1.21.1 (67.1 g) in 300 mL acetonitrile, and the mixture was heated to 70 oC overnight. The mixture was cooled, poured into diethyl ether, and rinsed with water and brine. The mixture was concentrated and chromatographed on silica gel using 2-20% ethyl acetate in heptane to give the title compound.
1.21.3 methyl 3-(2-aminoethyl)-5-bromobenzoate
Borane-tetrahydrofuran complex (126 mL, 1M solution) was added to a solution of Example 1.21.2 (16 g) in 200 mL tetrahydrofuran, and the mixture was stirred overnight. The reaction was carefully quenched with methanol (50 mL), and then concentrated to 50 mL volume. The mixture was then taken up in 120 mL methanol / 120 mL 4M HCl / 120 mL dioxane, and stirred overnight. The organics were removed by evaporation under reduced pressure, and the residue was extracted with diethyl ether (2 x). The organic extracts were discarded. The aqueous layer was basified with solid K2CO3, and then extracted with ethyl acetate, and dichloromethane (2x). The extracts were combined, dried over Na2SO4, filtered and concentrated to give the title compound.
1.21.4 methyl 3-bromo-5-(2-(2,2,2-trifluoroacetamido)ethyl)benzoate Trifluoroacetic anhydride (9.52 mL) was added dropwise to a mixture of Example 1.21.3 (14.5 g) and triethylamine (11.74 mL) in 200 mL dichloromethane at 0 °C. Upon addition, the mixture was allowed to warm to room temperature and was stirred for three days. The mixture was poured into diethyl ether, and washed with NaHCO3 solution and brine. The mixture was concentrated and chromatographed on silica gel using 5-30% ethyl acetate in heptanes to give the title compound.
1.21.5 methyl 6-bromo-2-(2,2,2-trifluoroacetyl)-1,2,3,4- tetrahydroisoquinoline-8-carboxylate
Sulfuric acid was added to Example 1.21.4 (10 g) until it went into solution (40 mL), at which time paraformaldehyde (4.24 g) was added, and the mixture was stirred for 2 hours. The solution was then poured onto 400 mL ice, and stirred 10 minutes. It was then extracted with ethyl acetate (3x), and the combined extracts were washed with NaHCO3 solution and brine, and then concentrated The crude product was chromatographed on silica gel using 2-15% ethyl acetate in heptanes to give the title compound. 1.21.6methyl 6-((2-((tert-butoxycarbonyl)(methyl)amino)ethyl)(methyl)amino)-2- (2,2,2-trifluoroacetyl)-1,2,3,4-tetrahydroisoquinoline-8-carboxylate Example 1.21.5 (2.25 g), tert-butyl methyl(2-(methylamino)ethyl)carbamate (1.27 g), palladium (II) acetate (0.083 g), 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene (0.213 g) and cesium carbonate (4.00 g) were stirred in 40 mL dioxane at 80 oC overnight. The mixture was concentrated and chromatographed on silica gel using 5-50% ethyl acetate in heptanes to give the title compound.
1.21.7 methyl 2-(5-bromo-6-(tert-butoxycarbonyl)pyridin-2-yl)-6-((2-((tert- butoxycarbonyl)(methyl)amino)ethyl)(methyl)amino)-1,2,3,4- tetrahydroisoquinoline-8-carboxylate
Example 1.21.6 (3 g) and potassium carbonate (2.63 g) were stirred in 30 mL tetrahydrofuran, 20 mL methanol, and 25 mL water overnight. The mixture was concentrated and 60 mL N,N- dimethylformamide was added. To this was then added Example 1.4.4 (1.08 g) and triethylamine (0.6 mL), and the reaction was stirred at 50 oC overnight. The mixture was cooled to room temperature and poured into ethyl acetate (200 mL). The solution was washed with water (3x) and brine, then dried over Na2SO4, filtered, and concentrated. The residue was chromatographed on silica gel using 5-50% ethyl acetate in heptanes to give the title compound. MS (ESI) m/e 635 (M+H)+.
1.21.8 methyl 6-((2-((tert- butoxycarbonyl)(methyl)amino)ethyl)(methyl)amino)-2-(6-(tert- butoxycarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)pyridin-2-yl)-1,2,3,4-tetrahydroisoquinoline-8-carboxylate
The title compound was prepared by substituting Example 1.21.7 for Example 1.1.9 in Example 1.1.10.
1.21.9 methyl 6-((2-((tert- butoxycarbonyl)(methyl)amino)ethyl)(methyl)amino)-2-(6-(tert- butoxycarbonyl)-5-(1-((3-(2-methoxyethoxy)-5,7-dimethyladamantan- 1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxylate
The title compound was prepared by substituting Example 1.21.8 for Example 1.5.11 and Example 1.17.1 for Example 1.5.10 in Example 1.5.12. MS (ESI) m/e 885.6 (M+H)+.
1.21.10 6-((2-((tert-butoxycarbonyl)(methyl)amino)ethyl)(methyl)amino)-2-(6- (tert-butoxycarbonyl)-5-(1-((3-(2-methoxyethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2- yl)-1,2,3,4-tetrahydroisoquinoline-8-carboxylic acid
The title compound was prepared by substituting Example 1.21.9 for Example 1.4.7 in Example 1.4.8. 1.21.11 tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-6-((2-((tert- butoxycarbonyl)(methyl)amino)ethyl)(methyl)amino)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-methoxyethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate The title compound was prepared by substituting Example 1.21.10 for Example 1.4.8 in Example 1.4.9. MS (ESI) m/e 1003.6 (M+H)+.
1.21.12 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-6-{methyl[2- (methylamino)ethyl]amino}-3,4-dihydroisoquinolin-2(1H)-yl]-3-(1-{[3- (2-methoxyethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5- methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid
Example 1.21.11 (40 mg) was stirred in 2 mL trifluoroacetic acid and 3 mL dichloromethane overnight. After evaporation of the solvent, the residue was purified on an HPLC (Gilson system, eluting with 10-85% acetonitrile in 0.1% trifluoroacetic acid in water) to give the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 12.75 (bs, 1H), 12.50 (br s, 1H), 8.40 (m, 2H), 8.01 (d, 1H), 7.76 (d, 1H), 7.45 (m, 2H), 7.32 (t, 1H), 7.24 (s, 1H), 6.99 (d, 1H), 6.86 (d, 1H), 6,78 (d, 1H), 4.72 (m, 2H), 3.98 (m, 2H), 3.80 (m, 4H), 3.76 (s, 2H), 3.55 (m, 2H), 3.29 (d, 3H), 3.20 (s, 3H), 3.15 (m, 2H), 2.90 (s, 3H), 2.58 (t, 2H), 2.05 (s, 3H), 1.30 (s, 2H), 1.21 (m, 4H), 1.08 (m, 4H), 0.98 (m, 2H), 0.85 (s, 6H). MS (ESI) m/e 847.5 (M+H)+.
1.22 Synthesis of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-6-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol- 4-yl]pyridine-2-carboxylic acid (W3.22)
1.22.1 methyl 6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-(2,2,2- trifluoroacetyl)-1,2,3,4-tetrahydroisoquinoline-8-carboxylate
A mixture of Example 1.21.5 (4.5 g), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2- dioxaborolane) (3.75 g), [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II)
dichloromethane (0.4 g), and potassium acetate (3.62 g) was stirred in 60 mL dioxane at 70 oC for 24 hours. The mixture was then diluted with ethyl acetate, and rinsed with water and brine. The mixture was concentrated and chromatographed on silica gel using 5-50% ethyl acetate in heptanes to give the title compound.
1.22.2 methyl 6-hydroxy-2-(2,2,2-trifluoroacetyl)-1,2,3,4- tetrahydroisoquinoline-8-carboxylate
Hydrogen peroxide (30%, 1.1 mL) was added to a mixture of Example 1.22.1 (4 g) and 1M aqueous NaOH solution (9.86 mL) in 40 mL tetrahydrofuran and 40 mL water, and the mixture was stirred for 90 minutes. The solution was acidified with concentrated HCl, and extracted twice with ethyl acetate. The combined extracts were washed with brine. The mixture was then concentrated and chromatographed on silica gel using 5-50% ethyl acetate in heptanes to give the title compound. MS (ESI) m/e 304.2 (M+H)+.
1.22.3 methyl 6-methoxy-2-(2,2,2-trifluoroacetyl)-1,2,3,4- tetrahydroisoquinoline-8-carboxylate
Trimethylsilyldiazomethane (2.6 mL, 2M solution in diethyl ether) was added to Example 1.22.2 (800 mg) in 10 mL methanol, and the reaction was stirred for 24 hours. The mixture was then concentrated and chromatographed on silica gel using 5-25% ethyl acetate in heptanes to give the title compound. MS (ESI) m/e 318.2 (M+H)+.
1.22.4 methyl 2-(5-bromo-6-(tert-butoxycarbonyl)pyridin-2-yl)-6-methoxy- 1,2,3,4-tetrahydroisoquinoline-8-carboxylate
The title compound was prepared by substituting Example 1.22.3 for Example 1.21.6 in Example 1.21.7. MS (ESI) m/e 479.1 (M+H)+.
1.22.5 methyl 2-(6-(tert-butoxycarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyridin-2-yl)-6-methoxy-1,2,3,4- tetrahydroisoquinoline-8-carboxylate
The title compound was prepared by substituting Example 1.22.4 for Example 1.1.9 in Example 1.1.10. MS (ESI) m/e 525.1 (M+H)+.
1.22.6 methyl 2-(6-(tert-butoxycarbonyl)-5-(1-((-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-6-methoxy-1,2,3,4- tetrahydroisoquinoline-8-carboxylate
The title compound was prepared by substituting Example 1.22.5 for Example 1.5.11 and Example 1.1.9 for Example 1.5.10 in Example 1.5.12. MS (ESI) m/e 829.6 (M+H)+.
1.22.7 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-6-methoxy-1,2,3,4- tetrahydroisoquinoline-8-carboxylic acid
The title compound was prepared by substituting Example 1.22.6 for Example 1.4.7 in Example 1.4.8. MS (ESI) m/e 814.6 (M+H)+.
1.22.8 tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-6-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate
The title compound was prepared by substituting Example 1.22.7 for Example 1.4.8 in Example 1.4.9. MS (ESI) m/e 946.5 (M+H)+.
1.22.9 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-6-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-3-[1-({35-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl- 1H-pyrazol-4-yl]pyridine-2-carboxylic acid
The title compound was prepared by substituting Example 1.22.8 for Example 1.21.11 in Example 1.21.12. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 12.75 (bs, 1H), 12.50 (br s, 1H), 8.21 (m, 2H), 8.01 (d, 1H), 7.76 (d, 1H), 7.44 (m, 2H), 7.32 (t, 1H), 7.25 (s, 1H), 7.20 (d, 1H), 6.99 (d, 1H), 6.90 (d, 1H), 4.72 (m, 2H), 3.80 (m, 4H), 3.55 (s, 3H), 3.50 (d, 3H), 2.98 (m, 4H), 2.51 (t, 2H), 2.05 (s, 3H), 1.35 (s, 2H), 1.26 (m, 4H), 1.10 (m, 4H), 1.00 (m, 2H), 0.85 (s, 6H). MS (ESI) m/e 790.4 (M+H)+.
1.23 Synthesis of 3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- y]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[4-(1,3-benzothiazol-2- ylcarbamoyl)quinolin-6-yl]pyridine-2-carboxylic acid (W3.23)
1.23.1 ethyl 6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoline-4- carboxylate
To a solution of ethyl 6-bromoquinoline-4-carboxylate (140 mg) in N,N-dimethylformamide (2 mL) was added [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II) dichloromethane (20 mg), potassium acetate (147 mg) and bis(pinacolato)diboron (190 mg). The mixture was stirred at 60 °C overnight. The mixture was cooled to room temperature and used in the next reaction directly. MS (ESI) m/e 328.1 (M+H)+.
1.23.2di-tert-butyl {2-[(3,5-dimethyl-7-{[5-methyl-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-1H-pyrazol-1-yl]methyl}tricyclo[3.3.1.13,7]decan-1- yl)oxy]ethyl}-2-imidodicarbonate
To a solution of Example 1.20.3 (13 g) in dioxane (100 mL) was added dicyclohexyl(2',6'- dimethoxy-[1,1'-biphenyl]-2-yl)phosphine (S-Phos) (1.0 g) and bis(benzonitrile)palladium(II) chloride (0.23 g) and the reaction was purged with several house vacuum/N2 refills. 4,4,5,5-Tetramethyl- 1,3,2-dioxaborolane (8.8 mL) and triethylamine (8.4 mL) was added followed by a couple more house vacuum/nitrogen refills and then the reaction was heated to 85 °C under nitrogen for 90 minutes. The reaction was cooled, filtered through diatomaceous earth and rinsed with methyl tert-butyl ether. The solution was then concentrated and chromatographed on silica gel using 25% ethyl acetate in heptanes to give the title compound.
1.23.3 tert-butyl 3-{1-[(3-{2-[bis(tert-butoxycarbonyl)amino]ethoxy}-5,7- dimethyltricyclo[3.3.1.13,7]decan-1-yl)methyl]-5-methyl-1H-pyrazol-4- yl}-6-chloropyridine-2-carboxylate
To a solution of Example 1.23.2 (12.3 g) and tert-butyl 3-bromo-6-chloropicolinate (5.9 g) in dioxane (50 mL) was added (1S,3R,5R,7S)-1,3,5,7-tetramethyl-8-phenyl-2,4,6-trioxa-8- phosphaadamantane(CyTop) (0.52 g) and bis(dibenzylideneacetone)palladium(0) (0.66 g). After several house vacuum/nitrogen refills, potassium phosphate (4.06 g) and water (25 mL) were added and the reaction was heated at 80 °C under nitrogen for 30 minutes. The reaction was cooled and then water and ethyl acetate were added. The organic layer was separated and washed with brine. The combined aqueous layers were extracted with ethyl acetate, and dried over sodium sulfate. The solution was filtered, concentrated and chromatographed on silica gel using 33% ethyl acetate in heptanes to give the title compound.
1.23.4 ethyl 6-[5-{1-[(3-{2-[bis(tert-butoxycarbonyl)amino]ethoxy}-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol-4- yl}-6-(tert-butoxycarbonyl)pyridin-2-yl]quinoline-4-carboxylate
To a solution of Example 1.23.1 (164 mg) in 1,4-dioxane (10 mL) and water (5 mL) was added Example 1.23.3 (365 mg), bis(triphenylphosphine)palladium(II) dichloride (35 mg), and CsF (228 mg). The mixture was stirred at 120 oC for 30 minutes under microwave conditions (Biotage Initiator). The mixture was diluted with ethyl acetate (200 mL) and washed with water and brine and dried over anhydrous sodium sulfate. Filtration and evaporation of the solvent gave a residue that purified by silica gel chromatography, eluting with 20% ethyl acetate in heptane, to give the title compound. MS (ESI) m/e 894.3(M+H)+.
1.23.5 6-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)amino)ethoxy)-5,7-dimethyladamantan-1-yl)methyl)- 5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)quinoline-4-carboxylic acid To a solution of Example 1.23.4 (3.1 g) in tetrahydrofuran (20 mL), methanol (10 mL) and water (10 mL) was added LiOH H2O (240 mg). The mixture was stirred at room temperature overnight. The mixture was acidified with aqueous 2N HCl, diluted with ethyl acetate (400 mL), washed with water and brine, and dried over anhydrous sodium sulfate. Filtration and evaporation of the solvent gave the title compound, which was used without further purification. MS (ESI) m/e 766.3(M+H)+.
1.23.6 3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[4-(1,3-benzothiazol-2- ylcarbamoyl)quinolin-6-yl]pyridine-2-carboxylic acid
To a solution of Example 1.23.5 (4.2 g) in dichloromethane (30 mL) was added
benzo[d]thiazol-2-amine (728 mg), 1-ethyl-3-[3-(dimethylamino)propyl]-carbodiimide hydrochloride (1.40 g) and 4-(dimethylamino)pyridine (890 mg). The mixture was stirred at room temperature overnight. The reaction mixture was diluted with ethyl acetate (500 mL), washed with water and brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was dissolved in dichloromethane and trifluoroacetic acid (10 mL, 1:1) and stirred overnight. The solvents were removed under reduced pressure. The residue was diluted with N,N- dimethylformamide (2 mL), filtered and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1% trifluoroacetic acid, to give the title compound 1H NMR (400 MHz dimethyl sulfoxide-d6) δ ppm 912 (dd 1H) 892 (s 1H) 861 (dt, 1H), 8.35– 8.16 (m, 2H), 8.07 (d, 1H), 7.97– 7.87 (m, 2H), 7.81 (d, 1H), 7.66 (s, 3H), 7.53– 7.44 (m, 2H), 7.38 (t, 1H), 3.88 (s, 2H), 3.49 (t, 2H), 2.89 (q, 2H), 2.22 (s, 4H), 1.43 (s, 2H), 1.29 (q, 4H), 1.15 (s, 4H), 1.09– 0.96 (m, 2H), 0.86 (s, 7H). MS (ESI) m/e 742.2 (M+H)+.
1.24 Synthesis of 6-[5-amino-8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol- 4-yl]pyridine-2-carboxylic acid (W3.24)
1.24.1 5-tert-butoxycarbonylamino-2-(2,2,2-trifluoro-acetyl)-1,2,3,4- tetrahydro-isoquinoline-8-carboxylic acid methyl ester
Example 1.13.4 (5000 mg), tert-butyl carbamate (1920 mg), and cesium carbonate (6674 mg) were added to 1,4-dioxane (80 mL). The solution was degassed and flushed with nitrogen three times. Diacetoxypalladium (307 mg) and (9,9-dimethyl-9H-xanthene-4,5-diyl)bis(diphenylphosphine) (1580 mg) were added, and the solution was degassed and flushed with nitrogen once. The solution was heated to 80 °C for 16 hours. The solution was cooled, and 1 M aqueous HCl (150 mL) was added. The solution was extracted with 50% ethyl acetate in heptanes. The organic portion was washed with brine and dried on anhydrous sodium sulfate. The solution was filtered, concentrated and purified by flash column chromatography on silica gel, eluting with 30% ethyl acetate in heptanes. The solvent was removed under reduced pressure to yield the title compound. MS (ESI) m/e 420 (M+NH4)+, 401 (M-H)-.
1.24.2 5-tert-butoxycarbonylamino-1,2,3,4-tetrahydro-isoquinoline-8- carboxylic acid methyl ester
The title compound was prepared by substituting Example 1.24.1 for Example 1.13.5 in Example 1.13.6. MS (ESI) m/e 307 (M+H)+, 305 (M-H)-.
1.24.3 2-(5-bromo-6-tert-butoxycarbonyl-pyridin-2-yl)-5-tert- butoxycarbonylamino-1,2,3,4-tetrahydro-isoquinoline-8-carboxylic acid methyl ester
The title compound was prepared by substituting Example 1.24.2 for Example 1.13.6 in Example 1.13.7. MS (ESI) m/e 562, 560 (M+H)+, 560, 558 (M-H)-.
1.24.4 5-tert-butoxycarbonylamino-2-[6-tert-butoxycarbonyl-5-(4,4,5,5- tetramethyl-[1,3,2]dioxaborolan-2-yl)-pyridin-2-yl]-1,2,3,4-tetrahydro- isoquinoline-8-carboxylic acid methyl ester
The title compound was prepared by substituting Example 1.24.3 for Example 1.13.7 in Example 1.13.8. MS (ESI) m/e 610 (M+H)+, 608 (M-H)-.
1.24.5 methyl 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-5-((tert- butoxycarbonyl)amino)-1234-tetrahydroisoquinoline-8-carboxylate The title compound was prepared by substituting Example 1.24.4 for Example 1.13.8 and Example 1.1.9 for Example 1.4.2 in Example 1.13.9. MS (ESI) m/e 913 (M+H)+, 911 (M-H)-.
1.24.6 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-5-((tert- butoxycarbonyl)amino)-1,2,3,4-tetrahydroisoquinoline-8-carboxylic acid
The title compound was prepared by substituting Example 1.24.5 for Example 1.13.9 in Example 1.13.10. MS (ESI) m/e 899 (M+H)+, 897 (M-H)-.
1.24.7 tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-((tert- butoxycarbonyl)amino)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2- ((tert-butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan- 1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate
The title compound was prepared by substituting Example 1.24.6 for Example 1.13.10 in Example 1.13.11. MS (ESI) m/e 1031 (M+H)+, 1029 (M-H)-.
1.24.8 6-[5-amino-8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl- 1H-pyrazol-4-yl]pyridine-2-carboxylic acid
The title compound was prepared by substituting Example 1.24.7 for Example 1.13.11 in Example 1.13.12. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 11.42 (s, 1H), 7.98 (d, 1H), 7.75 (d, 1H), 7.55 (d, 1H), 7.44 (t, 2H), 7.31 (t, 1H), 7.27 (s, 1H), 6.92 (d, 1H), 6.58 (d, 1H), 5.74 (s, 2H), 4.99 (s, 2H), 3.93 (t, 2H), 3.82 (s, 2H), 3.57 (s, 3H), , 3.54 (m, 2H), 3.09 (q, 2H), 2.98 (bs, 2H), 2.11 (s, 3H), 1.35-1.04 (m, 12H), 0.87 (s, 6H). MS (ESI) m/e 775 (M+H)+.
1.25 Synthesis of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-6-[3- (methylamino)prop-1-yn-1-yl]-3,4-dihydroisoquinolin-2(1H)-yl]-3-(1-{[3-(2- methoxyethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)pyridine-2-carboxylic acid (W3.25)
1.25.1 methyl 6-(3-((tert-butoxycarbonyl)(methyl)amino)prop-1-yn-1- yl)-2-(2,2,2-trifluoroacetyl)-1,2,3,4-tetrahydroisoquinoline-8- carboxylate
A solution of Example 1.21.5 (1.97 g), tert-butyl methyl(prop-2-yn-1-yl)carbamate (1 g), bis(triphenylphosphine)palladium(II) dichloride (0.19 g), CuI (0.041 g), and triethylamine (2.25 mL) in 20 mL dioxane was stirred at 50 oC overnight. The mixture was then concentrated and
chromatographed on silica gel using 10-50% ethyl acetate in heptanes to give the title compound. 1.25.2 methyl 2-(5-bromo-6-(tert-butoxycarbonyl)pyridin-2-yl)-6-(3- ((tert-butoxycarbonyl)(methyl)amino)prop-1-yn-1-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxylate
The title compound was prepared by substituting Example 1.25.1 for Example 1.21.6 in Example 1.21.7. MS (ESI) m/e 616 (M+H)+.
1.25.3 methyl 6-(3-((tert-butoxycarbonyl)(methyl)amino)prop-1-yn-1- yl)-2-(6-(tert-butoxycarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyridin-2-yl)-1,2,3,4-tetrahydroisoquinoline-8- carboxylate
The title compound was prepared by substituting Example 1.25.2 for Example 1.1.9 in Example 1.1.10. MS (ESI) m/e 662.3 (M+H)+.
1.25.4 methyl 6-(3-((tert-butoxycarbonyl)(methyl)amino)prop-1-yn-1- yl)-2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-methoxyethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2- yl)-1,2,3,4-tetrahydroisoquinoline-8-carboxylate
The title compound was prepared by substituting Example 1.25.3 for Example 1.5.11 and Example 1.17.1 for Example 1.5.10 in Example 1.5.12.
1.25.5 6-(3-((tert-butoxycarbonyl)(methyl)amino)prop-1-yn-1-yl)-2-(6- (tert-butoxycarbonyl)-5-(1-((3-(2-methoxyethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2- yl)-1,2,3,4-tetrahydroisoquinoline-8-carboxylic acid
The title compound was prepared by substituting Example 1.25.4 for Example 1.4.7 in Example 1.4.8.
1.25.6 tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-6-(3-((tert- butoxycarbonyl)(methyl)amino)prop-1-yn-1-yl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-methoxyethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate The title compound was prepared by substituting Example 1.25.5 for Example 1.4.8 in Example 1.4.9.
1.25.7 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-6-[3-(methylamino)prop- 1-yn-1-yl]-3,4-dihydroisoquinolin-2(1H)-yl]-3-(1-{[3-(2- methoxyethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5- methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid
The title compound was prepared by substituting Example 1.25.6 for Example 1.21.11 in Example 1.21.12. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 12.95 (bs, 1H), 8.70 (m, 1H), 8.02 (d, 1H), 7.77 (d, 1H), 7.74 (m, 1H), 7.47 (m, 2H), 7.34 (m, 2H), 7.24 (s, 1H), 6.95 (m, 1H), 6.78 (m, 1H), 4.92 (s, 2H), 4.28 (t, 2H), 3.95 (t, 2H), 3.40 (s, 3H), 3.30 (m, 2H), 3.20 (s, 3H), 3.00 (m, 2H), 2.57 (t, 2H), 2.07 (s, 3H), 1.85 (m, 2H), 1.29 (d, 2H), 1.10-1.24 (m, 10H), 0.85 (s, 6H).
1.26 Synthesis of 6-[4-(1,3-benzothiazol-2-ylcarbamoyl)isoquinolin-6-yl]-3-[1- ({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5- methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid (W3.26)
1.26.1 methyl 2-(3-bromophenyl)-2-cyanoacetate
To a solution of 2-(3-bromophenyl)acetonitrile (5 g) in tetrahydrofuran (50 mL) was added sodium hydride (3.00 g) portion wise at 23°C. The mixture was heated to 50 °C for 20 minutes. Dimethyl carbonate (8.60 mL) was added dropwise. The mixture was heated at reflux for 2 hours. The mixture was poured into cold and slightly acidic water. The aqueous layer was extracted with ethyl acetate (2 x 200 mL). The combined organic layers were washed with brine, dried over anhydrous sodium sulfate, filtered through a Büchner funnel and concentrated to give a residue, which was purified by silica gel column chromatography, eluting with 0%-25% dichloromethane/ petroleum ether to afford the title compound. MS (LC-MS) m/e 256.0 (M+H) +
1.26.2 methyl 3-amino-2-(3-bromophenyl)propanoate
Sodium borohydride (14.89 g, 394 mmol) was added portionwise to a solution of Example 1.26.1 (10 g) and cobalt(II) chloride hexahydrate (18.73 g) in methanol (200 mL) at -20 °C. The mixture was stirred for 1 hour and the pH was adjusted to 3 with 2N aqueous HCl. The mixture was concentrated. The residue was basified with 2 M aqueous sodium hydroxide and extracted with ethyl acetate. The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated to provide the title compound. MS (LC-MS) m/e 260.0 (M+H) +.
1.26.3 methyl 2-(3-bromophenyl)-3-formamidopropanoate A solution of Example 1.26.2 (3.6 g) in ethyl formate (54 mL) was heated at 80 °C for 5 hours. The solvent was removed, and the residue was purified by silica gel column chromatography eluting with petroleum/ethyl acetate (2:1-1:2) to give the title compound. MS (LC-MS) m/e 288.0 (M+H)+.
1.26.4 methyl 8-bromo-2,3-dioxo-3,5,6,10b-tetrahydro-2H-oxazolo[2,3- a]isoquinoline-6-carboxylate
Oxalyl chloride (1.901 mL) was slowly added to a solution of Example 1.26.3 (5.65 g) in dichloromethane (190 mL). The resulting mixture was stirred at 20 °C for 2 hours. The mixture was cooled to -20 °C, and iron(III) chloride (3.84 g) was added. The resulting mixture was stirred at 20 °C for 3 hours. Aqueous hydrochloric acid (2M, 45 mL) was added in one portion, and the resulting biphasic mixture was vigorously stirred for 0.5 hours at room temperature. The biphasic mixture was poured into a separatory funnel, and the phases were separated. The organic layer was washed with brine, dried with sodium sulfate, and filtered. The solvent was evaporated under reduced pressure to provide the title compound. The crude product was directly used in subsequent step without purification. MS (LC-MS) m/e 342.0 (M+H) +. 1.26.5 methyl 6-bromo-3,4-dihydroisoquinoline-4-carboxylate Example 1.26.4 (13.0 g) in methanol (345 mL) and sulfuric acid (23 mL) was heated at 80 °C for 16 hours. The mixture was concentrated, and the residue was diluted with water, basified with saturated aqueous sodium bicarbonate solution and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate, filtered and
concentrated. The residue was purified by silica gel column chromatography, eluting with petroleum ether/ ethyl acetate (2:1-1:2) to give the title compound. MS (LC-MS) m/e 268.0 (M+H) +.
1.26.6 methyl 6-bromoisoquinoline-4-carboxylate
To a solution of Example 1.26.5 (5.25 g) in 1,4-dioxane (200 mL) at 60 °C was added manganese(IV) dioxide (8.5 g). The mixture was heated to 110 °C for 3 hours. The reaction mixture was filtered through a pad of diatomaceous earth and washed with dichloromethane and ethyl acetate. The filtrate was concentrated to dryness. The crude material was adsorbed onto silica gel and purified by silica gel chromatography, eluting with 5-30% ethyl acetate in dichloromethane to give the title compound. MS (LC-MS) m/e 267.9 (M+H) +.
1.26.7 methyl 6-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)isoquinoline-4- carboxylate
Example 1.26.6 (229 mg), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (328 mg) and potassium acetate (253 mg) in N,N-dimethylformamide (5 mL) was purged with N2 for 5 minutes and [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II) dichloromethane (42.2 mg) was added. The mixture was heated at 100 °C overnight and cooled. To the mixture was added Example 1.1.11 (0.369 g), dichlorobis(triphenylphosphine)palladium(II) (0.060 g), cesium fluoride (0.261 g) and water (2 mL). The resulting mixture was heated at 100 °C for 10 hours and filtered. The filtrate was concentrated. The residue was dissolved in dimethyl sulfoxide and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1% trifluoroacetic acid, to give the title compound. MS (ESI) m/e 794.5 (M+H)+.
1.26.8 6-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)isoquinoline-4- carboxylic acid
Example 1.26.7 (220 mg) in tetrahydrofuran-methanol was treated with 1 M aqueous sodium hydroxide (1.66 mL) for 2 days. The mixture was neutralized with acetic acid and concentrated. The residue was dissolved in dimethyl sulfoxide and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1% trifluoroacetic acid, to give the title compound. MS (ESI) m/e 780.5 (M+H)+. 1.26.9 tert-butyl 6-(4-(benzo[d]thiazol-2-ylcarbamoyl)isoquinolin-6-yl)-3-(1- ((3-(2-((tert-butoxycarbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate To a mixture of Example 1.26.8 (122 mg), benzo[d]thiazol-2-amine (47.0 mg), O-(7- azabenzotriazol-1-yl)-N,N,N’,N’-tetramethyluronium hexafluorophosphate (119 mg) in N,N- dimethylformamide (0.5 mL) was added N,N-diisopropylethylamine (273 µL). The mixture was stirred overnight and loaded onto an 80g silica gel column, eluting with 5-100% heptanes in ethyl acetate to provide the title compound. MS (ESI) m/e 912.5 (M+H)+.
1.26.10 6-[4-(1,3-benzothiazol-2-ylcarbamoyl)isoquinolin-6-yl]-3-[1-({3,5- dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid
Example 1.26.9 (100 mg) in dichloromethane (4 mL) was treated with trifluoroacetic acid (2 mL) for 3 hours and the mixture was concentrated. The residue was dissolved in dimethyl sulfoxide (5 mL) and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1% trifluoroacetic acid, to give the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ 13.27 (s, 1H), 9.58 (s, 1H), 9.03 (d, 2H), 8.53 (dd, 1H), 8.42 (d, 1H), 8.25 (t, 3H), 8.06 (d, 1H), 7.97 (d, 1H), 7.81 (d, 1H), 7.56– 7.45 (m, 2H), 7.37 (t, 1H), 3.89 (s, 2H), 3.55 (t, 2H), 3.01 (t, 2H), 2.54 (t, 4H), 2.23 (s, 3H), 1.44 (s, 2H), 1.36– 1.23 (m, 4H), 1.16 (s, 4H), 0.87 (s, 6H). MS (ESI) m/e 756.1 (M+H)+.
1.27 Synthesis of 6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-1H-indol-2-yl]-3-[1-({3,5- dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5- methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid (W3.27)
1.27.1 methyl 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-1H-indole-7- carboxylate
To a stirred solution of methyl 2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole-7- carboxylate (370 mg), tris(dibenzylideneacetone)dipalladium(0) (30 mg), 1,2,3,4,5-pentaphenyl-1′- (di-tert-butylphosphino)ferrocene (30 mg) and potassium phosphate (550 mg) in tetrahydrofuran (2 mL) was added Example 1.1.11 (735 mg). The mixture was purged with nitrogen and stirred at 70 °C for 3 hours. The reaction was diluted with ethyl acetate and washed with water and brine. The aqueous layer was back extracted by ethyl acetate. The combined organic layers were dried over sodium sulfate, filtered and concentrated. The residue was purified via silica gel chromatography, eluting with 0-20% ethyl acetate in heptanes, to give the title compound. MS (ESI) m/e 780.4 (M-H)-.
1.27.2 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-1H-indole-7- carboxylic acid
The title compound was prepared as described in Example 1.4.8, replacing Example 1.4.7 with Example 1.27.1. MS (ESI) m/e 766.4 (M-H)-.
1.27.3 tert-butyl 6-(7-(benzo[d]thiazol-2-ylcarbamoyl)-1H-indol-2-yl)-3-(1-((3- (2-((tert-butoxycarbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate The title compound was prepared as described in Example 1.4.9, replacing Example 1.4.8 with Example 1.27.2. MS (ESI) m/e 898.4 (M-H)\-.
1.27.4 6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-1H-indol-2-yl]-3-[1-({3,5- dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid
The title compound was prepared by substituting Example 1.27.3 for Example 1.1.13 in Example 1.1.14. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ ppm 13.01 (s, 1H), 11.19 (s, 1H), 8.27 (dd, 4H), 8.04 (d, 1H), 7.99 (d, 1H), 7.91 (d, 1H), 7.53– 7.45 (m, 3H), 7.36 (t, 1H), 7.27 (t, 1H), 3.91 (s, 2H), 3.57 (t, 3H), 3.03 (t, 3H), 2.58– 2.54 (m, 4H), 2.24 (s, 3H), 1.46 (s, 2H), 1.38– 1.27 (m, 4H), 1.24– 1.01 (m, 6H), 0.89 (s, 6H). MS (ESI) m/e 744.2 (M+H)+.
1.28 Synthesis of 3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-1H- indol-2-yl]pyridine-2-carboxylic acid (W3.28)
1.28.1 methyl 2-[5-{1-[(3-{2-[bis(tert-butoxycarbonyl)amino]ethoxy}-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol-4- yl}-6-(tert-butoxycarbonyl)pyridin-2-yl]-1H-indole-7-carboxylate
The title compound was prepared by substituting Example 1.23.3 for Example 1.1.11 in Example 1.27.1. MS (ESI) m/e 866.3 (M-H)-.
1.28.2 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)amino)ethoxy)-5,7-dimethyladamantan-1-yl)methyl)- 5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-1H-indole-7-carboxylic acid The title compound was prepared as described in Example 1.4.8, replacing Example 1.4.7 with Example 1.28.1. MS (ESI) m/e 754.4 (M+H)+.
1.28.3 tert-butyl 6-(7-(benzo[d]thiazol-2-ylcarbamoyl)-1H-indol-2-yl)-3-(1-((3- (2-((tert-butoxycarbonyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate
The title compound was prepared as described in Example 1.4.9, replacing Example 1.4.8 with Example 1.28.2. MS (ESI) m/e 886.5 (M+H)+. 1.28.4 3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[7-(1,3-benzothiazol-2- ylcarbamoyl)-1H-indol-2-yl]pyridine-2-carboxylic acid
The title compound was prepared by substituting Example 1.28.3 for Example 1.1.13 in Example 1.1.14. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ ppm 13.00 (s, 1H), 11.19 (s, 1H), 8.29 (d, 1H), 8.23 (d, 1H), 8.03 (d, 1H), 7.98 (d, 1H), 7.90 (d, 1H), 7.80 (s, 1H), 7.63 (s, 3H), 7.50 (s, 1H), 7.49– 7.44 (m, 2H), 7.39– 7.32 (m, 1H), 7.25 (t, 1H), 3.90 (s, 2H), 2.90 (q, 2H), 2.23 (s, 3H), 1.45 (s, 2H), 1.31 (q, 4H), 1.23– 1.00 (m, 7H), 0.88 (s, 6H). MS (ESI) m/e 730.2 (M+H)+.
1.29 Synthesis of 6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-3-methyl-1H-indol-2-yl]-3-[1- ({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5- methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid (W3.29)
1.29.1 methyl 3-methyl-1H-indole-7-carboxylate
To 7-bromo-3-methyl-1H-indole (1 g), dichloro[1,1'- bis(diphenylphosphino)ferrocene]palladium(II) dichloromethane adduct (0.070 g) in a 50 mL pressure bottle was added methanol (20 mL) and trimethylamine (1.327 mL). The reactor was purged with inert gas, followed by carbon monoxide. The reaction was heated to 100 °C for 20 hours at 60 psi. The solution was filtered and concentrated. The residue was purified by silica gel chromatography, eluting with a gradient of 5-30% ethyl acetate in heptanes, to give the title compound. MS (ESI) m/e 189.9 (M+H)+.
1.29.2 methyl 2-bromo-3-methyl-1H-indole-7-carboxylate
To a stirred suspension of Example 1.29.1 (70 mg) and 70 mg silica gel in dichloromethane (2 mL) was added 1-bromopyrrolidine-2,5-dione (70 mg). The mixture was protected from light by with aluminum foil and was stirred at room temperature under nitrogen for 30 minutes. The reaction mixture was filtered, washed with dichloromethane and purified via silica gel chromatography, eluting with 10-50% ethyl acetate in heptane, to provide the title compound. MS (ESI) m/e 267.6 (M+H)+.
1.29.3 methyl 3-methyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H- indole-7-carboxylate
To a stirred suspension of Example 1.29.2 (398 mg), 4,4,4',4',5,5,5',5'-octamethyl-2,2'- bi(1,3,2-dioxaborolane) (1.2 g,) and potassium acetate (450 mg) in 1,4-dioxane (2 mL) was added bis(triphenylphosphine)palladium(II) dichloride (55 mg). The mixture was purged with nitrogen and heated at 115 °C under microwave conditions (Biotage Initiator) for 3 hours. The reaction was diluted with ethyl acetate and washed with water and brine. The aqueous layer was back extracted with ethyl acetate. The combined organic layer was dried over sodium sulfate, filtered and concentrated. The residue was purified via silica gel chromatography, eluting with 5-50% ethyl acetate in heptane, to give the title compound. MS (ESI) m/e 315.9 (M+H)+. 1.29.4 methyl 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-3-methyl-1H-indole- 7-carboxylate
Example 1.29.4 was prepared by substituting Example 1.29.3 for methyl 2-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole-7-carboxylate in Example 1.27.1. MS (ESI) m/e 794.4 (M-H)-.
1.29.5 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-3-methyl-1H-indole- 7-carboxylic acid
Example 1.29.5 was prepared by substituting Example 1.29.4 for Example 1.4.7 in Example 1.4.8. MS (ESI) m/e 780.4 (M-H)-.
1.29.6 tert-butyl 6-(7-(benzo[d]thiazol-2-ylcarbamoyl)-3-methyl-1H-indol-2- yl)-3-(1-((3-(2-((tert-butoxycarbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate Example 1.29.6 was prepared by substituting Example 1.29.5 for Example 1.4.8 in Example 1.4.9. MS (ESI) m/e 912.4 (M-H)-.
1.29.7 6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-3-methyl-1H-indol-2-yl]-3-[1- ({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid
The title compound was prepared by substituting Example 1.29.6 for Example 1.1.13 in Example 1.1.14. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ ppm 12.97 (s, 1H), 11.04 (s, 1H), 8.34 – 8.23 (m, 3H), 8.06 (d, 1H), 8.02 (dd, 2H), 7.93 (d, 1H), 7.79 (d, 1H), 7.51 (s, 1H), 7.48 (ddd, 1H), 7.38– 7.32 (m, 1H), 7.25 (t, 1H), 3.91 (s, 2H), 3.56 (t, 2H), 3.03 (p, 2H), 2.67 (s, 3H), 2.56 (t, 3H), 2.25 (s, 3H), 1.46 (s, 2H), 1.38– 1.26 (m, 4H), 1.24– 1.13 (m, 4H), 1.06 (q, 2H), 0.89 (s, 6H). MS (ESI) m/e 758.2 (M+H)+.
1.30 Synthesis of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl]-3-(1-{[3,5-dimethyl-7-(2-{[1-(methylsulfonyl)piperidin-4- yl]amino}ethoxy)tricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H-pyrazol-4- yl)pyridine-2-carboxylic acid (W3.30)
1.30.1 tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-(((1r,7r)-3,5-dimethyl-7-(2-((1- (methylsulfonyl)piperidin-4-yl)amino)ethoxy)adamantan-1-yl)methyl)- 5-methyl-1H-pyrazol-4-yl)picolinate
A solution of Example 1.18.18 (0.060 g), 1-(methylsulfonyl)piperidin-4-one (0.015 g) and sodium triacetoxyborohydride (0024 g) was stirred in dichloromethane (05 mL) at room temperature After 30 minutes, the reaction mixture was concentrated. The crude material was dissolved in N,N- dimethylformamide (1.5 mL) and water (0.5 mL) and purified by preparatory reverse-phase HPLC on a Gilson 2020 system using a gradient of 5% to 85% acetonitrile/water. The product-containing fractions were lyophilized to give the title compound as a trifluoroacetic acid salt. MS (ESI) m/e 963.9 (M+H)+.
1.30.2 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)- yl]-3-(1-{[3,5-dimethyl-7-(2-{[1-(methylsulfonyl)piperidin-4- yl]amino}ethoxy)tricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)pyridine-2-carboxylic acid
A solution of Example 1.30.1 (0.060 g) was dissolved in dichloromethane (0.5 mL) and treated with trifluoroacetic acid (0.5 mL) overnight. The reaction mixture was concentrated. The residue was dissolved in N,N-dimethylformamide (1.5 mL) and water (0.5 mL) and was purified by preparatory reverse-phase HPLC on a Gilson 2020 system using a gradient of 5% to 85%
acetonitrile/water. The product-containing fractions were lyophilized to give the title compound.1H NMR (400 MHz, dimethyl sulfoxide-d6) δ 12.90 (s, 1H), 8.53 (d, 2H), 8.08 (d, 1H), 7.84 (d, 1H), 7.66 (d, 1H), 7.58– 7.45 (m, 4H), 7.41 (td, 2H), 7.33 (s, 1H), 7.00 (d, 1H), 5.00 (s, 2H), 3.93 (s, 2H), 3.88 (s, 2H), 3.62 (d, 4H), 3.22 (h, 2H), 3.12, 3.06 (s, 2H), 2.93 (s, 3H), 2.79 (d, 2H), 2.15 (s, 3H), 2.11 (s, 1H), 1.61 (qd, 2H), 1.48 (s, 2H), 1.37 (s, 2H), 1.19 (s, 4H), 1.10 (s, 2H), 0.91 (s, 8H). MS (ESI) m/e 907.2 (M+H)+.
1.31 Synthesis of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl]-3-(1-{[3,5-dimethyl-7-(2-{[1-(methylsulfonyl)azetidin-3- yl]amino}ethoxy)tricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H-pyrazol-4- yl)pyridine-2-carboxylic acid (W3.31)
A solution of Example 1.18.18 (0.050 g), 1-(methylsulfonyl)azetidin-3-one (0.014 g) and sodium triacetoxyborohydride (0.020 g) was stirred in dichloromethane (0.50 mL) at room temperature. After 30 minutes, acetic acid (5.35 µL) was added and stirring was continued at room temperature overnight. Trifluoroacetic acid (0.5 mL) was added to the reaction and was stirring continued overnight. The reaction mixture was concentrated. The residue was dissolved in a mixture of N,N-dimethylformamide (2 mL) and water (0.5 mL) and was purified by preparatory reverse-phase HPLC on a Gilson 2020 system using a gradient of 5% to 70% acetonitrile/water. The product- containing fractions were lyophilized to give the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ 12.86 (s, 1H), 9.13 (s, 2H), 8.03 (d, 1H), 7.79 (d, 1H), 7.62 (d, 1H), 7.54– 7.41 (m, 3H), 7.36 (td, 2H), 7.29 (s, 1H), 6.96 (d, 1H), 4.96 (s, 2H), 4.09 (s, 2H), 4.08 (s, 1H), 3.98 (s, 2H), 3.89 (s, 2H), 3.84 (s, 2H), 3.56 (s, 2H), 3.05 (s, 3H), 3.03 (s, 2H), 3.02 (s, 1H), 2.11 (s, 2H), 1.44 (s, 2H), 1.31 (q, 4H), 1.14 (s, 4H), 1.06 (s, 2H), 0.87 (s, 6H). MS (ESI) m/e 879.7 (M+H)+.
1.32 Synthesis of 3-{1-[(3-{2-[(3-amino-3-oxopropyl)amino]ethoxy}-5,7- dimethyltricyclo[331137]dec-1-yl)methyl]-5-methyl-1H-pyrazol-4-yl}-6-[8- (1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]pyridine-2- carboxylic acid (W3.32)
1.32.1 tert-butyl 3-(1-((3-(2-((3-amino-3-oxopropyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8- (benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)- yl)picolinate
A mixture of Example 1.18.18 (245 mg) and acrylamide (217 mg) in N,N-dimethylformamide (5 mL) was heated at 50 °C for 3 days and was purified by reverse phase HPLC, eluted with 30%- 80% acetonitrile in 0.1% trifluoroacetic acid in water solution, to provide the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ 12.83 (s, 1H), 8.30 (s, 2H), 8.00 (dd, 1H), 7.76 (d, 1H), 7.57 (d, 2H), 7.44 (ddd, 3H), 7.39– 7.29 (m, 2H), 7.21 (s, 1H), 7.13 (s, 1H), 6.91 (d, 1H), 4.95 (s, 2H), 3.81 (d, 4H), 3.53 (t, 2H), 3.05 (dq, 6H), 2.06 (s, 3H), 1.43 (s, 2H), 1.27 (q, 4H), 1.13 (d, 15H), 0.82 (s, 6H). MS (ESI) m/e 873.8 (M+H)+.
1.32.2 3-{1-[(3-{2-[(3-amino-3-oxopropyl)amino]ethoxy}-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol-4- yl}-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl]pyridine-2-carboxylic acid
The title compound was prepared using the procedure in Example 1.26.10, replacing Example 1.26.9 with Example 1.32.1. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ 8.29 (s, 2H), 8.00 (dd, 1H), 7.76 (d, 1H), 7.63– 7.52 (m, 2H), 7.49– 7.38 (m, 3H), 7.37– 7.29 (m, 2H), 7.25 (s, 1H), 7.11 (s, 1H), 6.92 (d, 1H), 4.92 (s, 2H), 3.53 (t, 2H), 3.04 (ddt, 6H), 2.07 (s, 3H), 1.39 (s, 2H), 1.26 (q, 4H), 1.16– 0.93 (m, 6H), 0.83 (s, 6H). MS (ESI) m/e 817.2 (M+H)+.
1.33 Synthesis of 6-[3-(1,3-benzothiazol-2-ylcarbamoyl)-1H-indazol-5-yl]-3-[1-({3,5- dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5- methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid (W3.33)
1.33.1 5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1-(2-trimethylsilanyl- ethoxymethyl)-1H-indazole-3-carboxylic acid ethyl ester
Ethyl 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indazole-3-carboxylate (1000 mg) was dissolved in N,N-dimethylformamide (30 mL). Sodium hydride (60% in mineral oil, 83 mg) was added, and the solution and was stirred at room temperature for 20 minutes. (2- (Chloromethoxy)ethyl)trimethylsilane (580 mg) was added, and the solution was stirred at room temperature for 90 minutes. The reaction was quenched with saturated aqueous ammonium chloride (10 mL) and diluted with water (90 mL). The solution was extracted with 70% ethyl acetate in heptanes (50 mL) twice. The combined organic portions were washed with water (25 mL) and then brine (25 mL). The solution was dried on anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by flash column chromatography on silica gel, eluting with 10-30% ethyl acetate in heptanes. The solvent was removed under reduced pressure to yield the title compound. MS (ESI) m/e 447 (M+H)+.
1.33.2 ethyl 5-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-1-((2- (trimethylsilyl)ethoxy)methyl)-1H-indazole-3-carboxylate
Example 1.33.1 (335 mg) and Example 1.1.11 (483 mg) were dissolved in 1,4-dioxane (3 mL). 2 M aqueous sodium carbonate (1.13 mL) was added, and the solution was degassed and flushed with nitrogen three times. Dichloro[1,1’-bis(diphenylphosphino)ferrocene]palladium(II) (61 mg) was added, and the solution was degassed and flushed with nitrogen once. The solution was heated at 75 °C for 16 hours. The solution was cooled, and 0.1 M aqueous HCl (25 mL) was added. The solution was extracted with ethyl acetate (50 mL) twice. The combined organic portions were washed with brine (25 mL) and dried on anhydrous sodium sulfate. The solution was filtered, concentrated under reduced pressure and purified by flash column chromatography on silica gel, eluting with 50% ethyl acetate in heptanes. The solvent was removed under reduced pressure to yield the title compound. MS (ESI) m/e 927 (M+NH4-H2O)+.
1.33.3 5-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-1-((2- (trimethylsilyl)ethoxy)methyl)-1H-indazole-3-carboxylic acid
The title compound was prepared by substituting Example 1.33.2 for Example 1.13.9 in Example 1.13.10. MS (ESI) m/e 899 (M+H)+, 897 (M-H)-.
1.33.4 tert-butyl 6-(3-(benzo[d]thiazol-2-ylcarbamoyl)-1-((2- (trimethylsilyl)ethoxy)methyl)-1H-indazol-5-yl)-3-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate
The title compound was prepared by substituting Example 1.33.3 for Example 1.13.10 in Example 1.13.11. MS (ESI) m/e 1030 (M+NH4-H2O)+, 1029 (M-H)-.
1.33.5 6-[3-(1,3-benzothiazol-2-ylcarbamoyl)-1H-indazol-5-yl]-3-[1-({3,5- dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid
Example 1.33.4 (83 mg) was dissolved in dichloromethane (0.5 mL). Trifluoroacetic acid (740 mg) was added, and the solution was stirred at room temperature for 16 hours. The solvents were removed under reduced pressure. The residue was dissolved in 1,4-dioxane (1 mL), and 1 M aqueous sodium hydroxide (0.5 mL) was added. The solution was stirred at room temperature for 60 minutes. The reaction was quenched with trifluoroacetic acid (0.1 mL) and purified by reverse-phase HPLC using 10-85% acetonitrile in water (w/01% trifluoroacetic acid) over 30 minutes on a Grace Reveleris® equipped with a Phenomenex® Luna® column: C18(2), 100 A, 150 x 30 mm. Product fractions were combined, frozen, and lyophilized to yield the title compound as the bis trifluoroacetic acid salt. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 14.23 (s, 1H), 12.58 (bs, 1H), 8.97 (s, 1H), 8.34-8.29 (m, 3H), 8.22 (d, 1H), 8.04 (d, 1H), 7.91 (d, 1H), 7.87-7.81 (m, 2H), 7.51-7.45 (m, 2H), 7.36 (t, 1H), 3.92 (s, 3H), 3.58 (m, 2H), 3.04 (m, 2H), 2.58-2.56 (m, 2H), 2.26 (s, 3H), 1.47 (s, 2H), 1.34 (q, 4H), 1.22-1.14 (m, 4H), 1.07 (q, 2H), 0.89 (m, 6H). MS (ESI) m/e 745 (M+H)+, 743 (M-H)-.
1.34 Synthesis of 6-[3-(1,3-benzothiazol-2-ylcarbamoyl)-1H-indol-5-yl]-3-[1-({3,5- dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5- methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid (W3.34)
1.34.1 5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1-(2-trimethylsilanyl- ethoxymethyl)-1H-indole-3-carboxylic acid methyl ester
The title compound was prepared by substituting methyl 5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-1H-indole-3-carboxylate for ethyl 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)-1H-indazole-3-carboxylate in Example 1.33.1. MS (ESI) m/e 432 (M+H)+.
1.34.2 methyl 5-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-1-((2- (trimethylsilyl)ethoxy)methyl)-1H-indole-3-carboxylate
The title compound was prepared by substituting Example 1.34.1 for Example 1.33.1 in Example 1.33.2. MS (ESI) m/e 912 (M+H)+.
1.34.3 5-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-1-((2- (trimethylsilyl)ethoxy)methyl)-1H-indole-3-carboxylic acid
The title compound was prepared by substituting Example 1.34.2 for Example 1.13.9 in Example 1.13.10. MS (ESI) m/e 898 (M+H)+, 896 (M-H)-.
1.34.4 tert-butyl 6-(3-(benzo[d]thiazol-2-ylcarbamoyl)-1-((2- (trimethylsilyl)ethoxy)methyl)-1H-indol-5-yl)-3-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate
The title compound was prepared by substituting Example 1.34.3 for Example 1.13.10 in Example 1.13.11. MS (ESI) m/e 1030 (M+H)+, 1028 (M-H)-.
1.34.5 6-[3-(1,3-benzothiazol-2-ylcarbamoyl)-1H-indol-5-yl]-3-[1-({3,5- dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid The title compound was prepared by substituting Example 1.34.4 for Example 1.33.4 in Example 1.33.5. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 12.47 (bs, 1H), 12.18 (s, 1H), 9.01 (s, 1H), 8.70 (d, 1H), 8.28 (bs, 3H), 8.12 (d, 1H), 8.05 (dd, 1H), 7.99 (d, 1H), 7.86 (d, 1H), 7.76 (d, 1H), 7.64 (d, 1H), 7.50 (s, 1H), 7.46 (td, 1H), 7.32 (t, 1H), 3.92 (s, 3H), 3.58 (m, 2H), 3.04 (m, 2H), 2.57 (m, 2H), 2.26 (s, 3H), 1.47 (s, 2H), 1.34 (q, 4H), 1.24-1.14 (m, 4H), 1.08 (m, 2H), 0.90 (s, 6H). MS (ESI) m/e 744 (M+H)+, 742 (M-H)-.
1.35 Synthesis of 6-[3-(1,3-benzothiazol-2-ylcarbamoyl)-1H-pyrrolo[2,3-b]pyridin-5- yl]-3-[1-({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid (W3.35) 1.35.1 5-bromo-1-(2-trimethylsilanyl-ethoxymethyl)-1H-pyrrolo[2,3- b]pyridine-3-carboxylic acid methyl ester
The title compound was prepared by substituting methyl 5-bromo-1H-pyrrolo[2,3-b]pyridine- 3-carboxylate for ethyl 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indazole-3-carboxylate in Example 1.33.1. MS (ESI) m/e 385, 387 (M+H)+.
1.35.2 5-(4,4,5,5-tetramethyl-[1,3,2]dioxaborolan-2-yl)-1-(2-trimethylsilanyl- ethoxymethyl)-1H-pyrrolo[2,3-b]pyridine-3-carboxylic acid methyl ester
The title compound was prepared by substituting Example 1.35.1 for Example 1.13.7 in Example 1.13.8. MS (ESI) m/e 433(M+H)+.
1.35.3 methyl 5-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-1-((2- (trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b]pyridine-3- carboxylate
The title compound was prepared by substituting Example 1.35.2 for Example 1.33.1 in Example 1.33.2. MS (ESI) m/e 913 (M+H)+.
1.35.4 5-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-1-((2- (trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b]pyridine-3-carboxylic acid
The title compound was prepared by substituting Example 1.35.3 for Example 1.13.9 in Example 1.13.10. MS (ESI) m/e 899 (M+H)+, 897 (M-H)-.
1.35.5 tert-butyl 6-(3-(benzo[d]thiazol-2-ylcarbamoyl)-1-((2- (trimethylsilyl)ethoxy)methyl)-1H-pyrrolo[2,3-b]pyridin-5-yl)-3-(1-((3- (2-((tert-butoxycarbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate The title compound was prepared by substituting Example 1.35.4 for Example 1.13.10 in Example 1.13.11. MS (ESI) m/e 1031 (M+H)+, 1029 (M-H)-.
1.35.6 6-[3-(1,3-benzothiazol-2-ylcarbamoyl)-1H-pyrrolo[2,3-b]pyridin-5-yl]- 3-[1-({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid
The title compound was prepared by substituting Example 1.35.5 for Example 1.33.4 in Example 1.33.5. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 12.74 (d, 1H), 12.62 (bs, 1H), 9.26 (d, 1H), 9.13 (d, 1H), 8.83 (d, 1H), 8.28 (bs, 2H), 8.25 (d, 1H), 7.99 (d, 1H), 7.91 (d, 1H), 7.78 (d, 1H), 7.51 (s, 1H), 7.47 (t, 1H), 7.33 (t, 1H), 3.92 (s, 3H), 3.58 (t, 2H), 3.04 (m, 2H), 2.57 (t, 2H), 2.26 (s, 3H), 1.47 (s, 2H), 1.34 (q, 4H), 1.20 (t, 4H), 1.08 (q, 2H), 0.90 (s, 6H). MS (ESI) m/e 745 (M+H)+, 743 (M-H)-.
1.36 Synthesis of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)- yl)-3-(1-((3-(2-((2-(N,N-dimethylsulfamoyl)ethyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinic acid (W3.36)
To a solution of Example 1.18.18 (69.8 mg) in N,N-dimethylformamide (6 mL) was added N,N-dimethylethenesulfonamide (118 mg), N,N-diisopropylethylamine (0.2 mL) and H2O (0.2 mL). The mixture was stirred at room temperature 4 days. The reaction mixture was diluted with ethyl acetate (200 mL), washed with water and brine, and dried over anhydrous sodium sulfate. After evaporation of the solvent, the residue was dissolved in dichloromethane and trifluoroacetic acid (10 mL, 1:1), and the resulting solution was stirred overnight. The solvents were removed under reduced pressure. The residue was diluted with N,N-dimethylformamide (2 mL), filtered and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1% trifluoroacetic acid, to give the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 12.82 (s, 1H), 8.53 (s, 2H), 8.00 (dd, 1H), 7.76 (d, 1H), 7.59 (dd, 1H), 7.53– 7.37 (m, 4H), 7.37– 7.28 (m, 2H), 7.26 (s, 1H), 6.92 (d, 1H), 4.92 (s, 2H), 3.80 (s, 2H), 3.54 (t, 2H), 3.44– 3.34 (m, 2H), 3.30 (s, 2H), 3.11 (s, 2H), 2.98 (t, 2H), 2.77 (s, 6H), 2.07 (s, 3H), 1.39 (s, 2H), 1.27 (q, 4H), 1.11 (s, 4H), 1.06– 0.93 (m, 2H), 0.83 (s, 7H). MS (ESI) m/e 881.2 (M+H)+.
1.37 Synthesis of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-3-{1-[(3-{2- [(3-hydroxypropyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl)methyl]-5-methyl-1H-pyrazol-4-yl}pyridine-2-carboxylic acid (W3.37) 1.37.1 2-((3,5-dimethyl-7-((5-methyl-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-1H-pyrazol-1-yl)methyl)adamantan-1- yl)oxy)ethanol
To a solution of Example 1.1.6 (8.9 g) and [1,1'- bis(diphenylphosphino)ferrocene]dichloropalladium(II) dichloromethane (818 mg) in acetonitrile (120 mL) was added triethylamine (10 mL) and pinacolborane (128 mL) The mixture was stirred at reflux overnight. The mixture was cooled to room temperature and used in the next reaction directly. MS (ESI) m/e 467.3 (M+Na)+.
1.37.2 tert-butyl 6-chloro-3-(1-((3-(2-hydroxyethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate To a solution of tert-butyl 3-bromo-6-chloropicolinate (6.52 g) in tetrahydrofuran (100 mL) and water (20 mL) was added Example 1.37.1 (9.90 g), (1S,3R,5R,7S)-1,3,5,7-tetramethyl-8- tetradecyl-2,4,6-trioxa-8-phosphaadamantane (0.732 g), tris(dibenzylideneacetone)dipalladium(0) (1.02 g), and potassium phosphate (23.64 g), and the mixture was stirred at reflux overnight. The solvents were removed under vacuum. The residue was dissolved in ethyl acetate (500 mL), washed with water and brine, and dried over anhydrous sodium sulfate. Filtration and evaporation of the solvent gave a residue that purified by silica gel chromatography, eluting with 20% ethyl acetate in heptane, to give the title compound. MS (ESI) m/e 530.3 (M+H)+.
1.37.3 tert-butyl 3-{1-[(3-{2-[bis(tert-butoxycarbonyl)amino]ethoxy}-5,7- dimethyltricyclo[3.3.1.13,7]decan-1-yl)methyl]-5-methyl-1H-pyrazol-4- yl}-6-chloropyridine-2-carboxylatetert-butyl 6-chloro-3-(1-((3,5- dimethyl-7-(2-((methylsulfonyl)oxy)ethoxy)adamantan-1-yl)methyl)-5- methyl-1H-pyrazol-4-yl)picolinate
To a cooled (0 °C) stirring solution of Example 1.37.2 (3.88 g) in dichloromethane (30 mL) and triethylamine (6 mL) was added methanesulfonyl chloride (2.52 g). The mixture was stirred at room temperature for 4 hours. The reaction mixture was diluted with ethyl acetate (400 mL), washed with water and brine, and dried over anhydrous sodium sulfate. Filtration and evaporation of the solvent gave the title compound, which was used in the next reaction without further purification. MS (ESI) m/e 608.1 (M+H)+.
1.37.4 tert-butyl 3-{1-[(3-{2-[bis(tert-butoxycarbonyl)amino]ethoxy}-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol-4- yl}-6-chloropyridine-2-carboxylate
To a solution of Example 1.37.3 (151 mg) in N,N-dimethylformamide (3 mL) was added di-t- butyl iminodicarboxylate (54 mg). The mixture was stirred at room temperature overnight. The reaction mixture was diluted with ethyl acetate (200 mL), washed with water and brine, and dried over anhydrous sodium sulfate. Filtration and evaporation of the solvent gave the title compound, which was used in the next step without further purification. MS (ESI) m/e 729.4 (M+H)+.
1.37.5 7-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)amino)ethoxy)-5,7-dimethyladamantan-1-yl)methyl)- 5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-1-naphthoic acid
To a solution of methyl 7-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-naphthoate (257 mg ) in 1,4-dioxane (10 mL) and water (5 mL) was added Example 1.37.4 (600 mg),
bis(triphenylphosphine)palladium(II) dichloride (578 mg) and cesium fluoride (375 mg) The mixture was stirred at 120 °C for 30 minutes under microwave conditions (Biotage Initiator). The mixture was diluted with ethyl acetate (200 mL), washed with water and brine, dried over anhydrous sodium sulfate, filtered and concentrated. Evaporation of the solvent gave a residue that purified by silica gel chromatography, eluting with 20% ethyl acetate in heptane, to give an intermediate di-ester. The residue was dissolved in tetrahydrofuran (10 mL), methanol (5 mL) and water (5 mL) and LiOH H2O (500 mg) was added. The mixture was stirred at room temperature overnight. The mixture was acidified with aqueous 2N HCl, dissolved in 400 mL of ethyl acetate, washed with water and brine and dried over anhydrous sodium sulfate. Filtration and evaporation of the solvent gave the title compound. MS (APCI) m/e 765.3 (M+H)+.
1.37.6 3-(1-((3-(2-aminoethoxy)-5,7-dimethyladamantan-1-yl)methyl)-5- methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)naphthalen-2-yl)picolinic acid
To a solution of Example 1.37.5 (500 mg) in dichloromethane (10 mL) was added benzo[d]thiazol-2-amine (98 mg), 1-ethyl-3-[3-(dimethylamino)propyl]-carbodiimide hydrochloride (251 mg) and 4-(dimethylamino)pyridine (160 mg). The mixture was stirred at room temperature overnight. The reaction mixture was diluted with ethyl acetate (400 mL), washed with water and brine, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was dissolved in dichloromethane and trifluoroacetic acid (10 mL, 1:1), and the solution was stirred overnight. The solvents were removed, and the residue was dissolved in N,N-dimethylformamide (12 mL) and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1% trifluoroacetic acid, to give the title compound. MS (ESI) m/e 741.2 (M+H)+.
1.37.7 3-((tert-butyldimethylsilyl)oxy)propanal
To a solution of dimethyl sulfoxide (2.5 mL) in dichloromethane (40 mL) at -78 oC was added oxalyl chloride (1.5 mL). The mixture was stirred 20 minutes at -78 °C, and a solution of (3- ((tert-butyldimethylsilyl)oxy)propan-1-ol (1.9 g) in dichloromethane (10 mL) was added by syringe. After 1 hour, triethylamine (5 mL) was added. The cooling bath was removed, and the reaction was stirred overnight. The reaction mixture was diluted with ethyl acetate (300 mL), washed with water and brine, and dried over anhydrous sodium sulfate. Filtration and evaporation of solvent gave the title compound. MS (DCI) m/e 206.0(M+NH4)+.
1.37.8 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-3-{1-[(3-{2-[(3- hydroxypropyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl)methyl]-5-methyl-1H-pyrazol-4-yl}pyridine-2-carboxylic acid
To a solution of Example 1.37.6 (125 mg) in dichloromethane (10 mL) was added Example 1.37.7 (32 mg). The mixture was stirred at room temperature for 1 hour, and NaBH(OAc)3 (107 mg) was added to the reaction mixture. The mixture was stirred at room temperature overnight. To the reaction mixture was added 2N aqueous sodium hydroxide (5 mL) and the reaction stirred for 4 hours. The mixture was neutralized with aqueous 2N HCl and extracted with ethyl acetate (100 mL x 3). The combined organic layers were washed with aqueous 2% HCl, water and brine and dried over anhydrous sodium sulfate. Filtration and evaporation of the solvent gave a residue that was purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1% trifluoroacetic acid, to give a solid. The residue was dissolved in tetrahydrofuran (6 mL) and tetrabutyl ammonium fluoride (1 M in tetrahydrofuran, 4 mL) was added. The mixture was stirred at room temperature for 2 hours, and the solvents were removed under vacuum. The residue was dissolved in dimethyl sulfoxide/methanol (1:1 , 12 mL) and was purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1% trifluoroacetic acid, to give the title compound. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ ppm 13.09 (s, 1H), 9.01 (s, 1H), 8.36 (dd, 1H), 8.20 (ddd, 5H), 8.09– 8.02 (m, 1H), 8.03– 7.95 (m, 1H), 7.92 (d, 1H), 7.80 (d, 1H), 7.69 (dd, 1H), 7.53– 7.43 (m, 2H), 7.36 (ddd, 1H), 3.89 (s, 2H), 3.56 (t, 2H), 3.47 (t, 2H), 3.10– 2.93 (m, 4H), 2.22 (s, 3H), 1.78– 1.68 (m, 2H), 1.44 (s, 2H), 1.30 (q, 4H), 1.20– 1.11 (m, 4H), 1.04 (q, 2H), 0.87 (s, 7H). MS (ESI) m/e 799.2 (M+H)+.
1.38 Synthesis of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl]-3-(1-{[3-(2-{[3-(dimethylamino)-3-oxopropyl]amino}ethoxy)-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H-pyrazol-4-yl)pyridine- 2-carboxylic acid (W3.38)
To a solution of Example 1.18.18 (55 mg) in N,N-dimethylformamide (6 mL) was added N,N-dimethylacrylamide (73.4 mg), N,N-diisopropylethylamine (0.2 mL) and water (0.2 mL). The mixture was stirred at room temperature 4 days. The reaction mixture was diluted with ethyl acetate (200 mL), washed with water and brine, and dried over anhydrous sodium sulfate. After filtration and evaporation of the solvent, the residue was dissolved in dichloromethane and trifluoroacetic acid (10 mL, 1:1). After stirring for 16 hours, the mixture was concentrated under reduced pressure. The residue was dissolved in N,N-dimethylformamide (8 mL) and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1%
trifluoroacetic acid, to give the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 12.84 (s, 1H), 8.22 (s, 3H), 8.02 (d, 1H), 7.78 (d, 1H), 7.60 (d, 1H), 7.55– 7.39 (m, 3H), 7.39– 7.30 (m, 2H), 7.27 (s, 1H), 6.94 (d, 1H), 4.94 (s, 2H), 3.87 (t, 2H), 3.81 (s, 2H), 3.55 (t, 2H), 3.20– 2.95 (m, 6H), 2.92 (s, 3H), 2.82 (s, 3H), 2.69 (q, 3H), 2.09 (s, 3H), 1.40 (s, 2H), 1.28 (q, 4H), 1.14 (d, 4H), 1.07– 0.94 (m, 2H), 0.85 (s, 8H). MS (ESI) m/e 845.3 (M+H)+.
1.39 Synthesis of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl]-3-(1-{[3,5-dimethyl-7-(2-{[3-(methylamino)-3- oxopropyl]amino}ethoxy)tricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)pyridine-2-carboxylic acid (W3.39)
The title compound was prepared as described in Example 1.38, by replacing N,N- dimethylacrylamide with N-methylacrylamide 1H NMR (501 MHz dimethyl sulfoxide-d6) δ ppm 12.84 (s, 1H), 8.32 (s, 2H), 8.08– 7.96 (m, 2H), 7.78 (d, 1H), 7.60 (d, 1H), 7.52– 7.40 (m, 3H), 7.39 – 7.30 (m, 2H), 7.27 (s, 1H), 6.94 (d, 1H), 4.94 (s, 2H), 3.87 (t, 2H), 3.81 (s, 2H), 3.12 (p, 2H), 3.01 (dt, 4H), 2.57 (d, 3H), 2.09 (s, 3H), 1.40 (s, 2H), 1.28 (q, 5H), 1.18– 1.07 (m, 4H), 1.02 (q, 2H), 0.85 (s, 7H). MS (ESI) m/e 831.3 (M+H)+.
1.40 Synthesis of 3-(1-{[3-(2-aminoacetamido)-5,7-dimethyltricyclo[3.3.1.13,7]decan- 1-yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-{8-[(1,3-benzothiazol-2-yl)carbamoyl]- 3,4-dihydroisoquinolin-2(1H)-yl}pyridine-2-carboxylic acid (W3.40) 1.40.1 1-((3-bromo-5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H- pyrazole
To a cooled (-30 °C) solution of Example 1.1.3 (500 mg) in tetrahydrofuran (30 mL) was added n-butyllithium (9.67 mL), and the mixture was stirred at -30 °C for 2 hours. Methyl iodide (1.934 mL) was added dropwise at -30 °C. After completion of the addition, the mixture was stirred at -30 °C for additional 2 hours. 1N aqueous HCl in ice water was added slowly, such that the temperature was maintained below 0 °C, until the pH reached 6. The mixture was stirred at room temperature for 10 minutes, and was diluted with ice-water (10 mL) and ethyl acetate (20 mL). The layers were separated, and the aqueous was extracted twice with ethyl acetate. The combined organic phases were washed with brine, dried over MgSO4, filtered and concentrated. The residue was purified by flash silica gel chromatography, eluting with 15/1 to 10/1petroleum/ethyl acetate, to give the title compound. MS (LC-MS) m/e 337, 339 (M+H)+.
1.40.2 1-(3,5-dimethyl-7-((5-methyl-1H-pyrazol-1-yl)methyl)adamantan-1- yl)urea
Example 1.40.1 (2.7 g) and urea (4.81 g) were mixed and stirred at 140 °C for 16 hours. The mixture was cooled to room temperature and suspended in methanol (200 mL x 2). The insoluble material was removed by filtration. The filtrate was concentrated to give the title compound. MS (LC-MS) m/e 317.3 (M+H) +.
1.40.3 3,5-dimethyl-7-((5-methyl-1H-pyrazol-1-yl)methyl)adamantan-1-amine To a solution of Example 1.40.2 (2.53 g) in 20% ethanol in water (20 mL) was added sodium hydroxide (12.79 g). The mixture was stirred at 120 oC for 16 hours and at 140 °C for another 16 hours. 6N Aqueous HCl was added until the pH reached 6. The mixture was concentrated, and the residue was suspended in methanol (200 mL). The insoluble material was filtered off. The filtrate was concentrated to give the title compound as an HCl salt. MS (LC-MS) m/e 273.9 (M+H)+.
1.40.4 tert-butyl (2-((3,5-dimethyl-7-((5-methyl-1H-pyrazol-1- yl)methyl)adamantan-1-yl)amino)-2-oxoethyl)carbamate
To a solution of Example 1.40.3 (2.16 g) in N,N-dimethylformamide (100 mL) was added triethylamine (3.30 mL), 2-((tert-butoxycarbonyl)amino)acetic acid (1.799 g) and O-(7- azabenzotriazol-1-yl)-N,N,N’,N’-tetramethyluronium hexafluorophosphate (3.90 g). The mixture was stirred at room temperature for 2 hours Water (40 mL) was added and the mixture was extracted with ethyl acetate (70 mL x 2). The combined organic phases were washed with brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography, eluting with 3/1 to 2/1 petroleum/ethyl acetate, to give the title compound. MS (LC-MS) m/e 430.8 (M+H)+.
1.40.5 tert-butyl (2-((3-((4-iodo-5-methyl-1H-pyrazol-1-yl)methyl)-5,7- dimethyladamantan-1-yl)amino)-2-oxoethyl)carbamate
To an ambient solution of Example 1.40.4 (1.7 g) in N,N-dimethylformamide (20 mL) was added N-iodosuccinimide (1.066 g) in portions, and the mixture was stirred at room temperature for 16 hours. Ice-water (10 mL) and saturated aqueous Na2S2O3 solution (10 mL) were added. The mixture was extracted with ethyl acetate (30 mL x 2). The combined organic phases were washed with brine, dried over sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography, eluting with 3/1 to 2/1 petroleum/ethyl acetate, to give the title compound. MS (LC- MS) m/e 556.6 (M+H)+ .
1.40.6 methyl 2-(5-bromo-6-(tert-butoxycarbonyl)pyridin-2-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxylate
To a solution of methyl 1,2,3,4-tetrahydroisoquinoline-8-carboxylate hydrochloride (12.37 g) and Example 1.4.4 (15 g) in dimethyl sulfoxide (100 mL) was added N,N-diisopropylethylamine (12 mL), and the mixture was stirred at 50 oC for 24 hours. The mixture was then diluted with ethyl acetate (500 mL) and washed with water and brine. The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel
chromatography, eluting with 20% ethyl acetate in hexane, to give the title compound. MS (ESI) m/e 448.4 (M+H)+.
1.40.7 methyl 2-(6-(tert-butoxycarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyridin-2-yl)-1,2,3,4-tetrahydroisoquinoline-8- carboxylate
To a solution of Example 1.40.6 (2.25 g) and [1,1'- bis(diphenylphosphino)ferrocene]dichloropalladium(II)(205 mg) in acetonitrile (30 mL) was added triethylamine (3 mL) and pinacolborane (2 mL), and the mixture was stirred at reflux for 3 hours. The mixture was diluted with ethyl acetate (200 mL) and washed with water and brine. The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure. Purification of the residue by flash chromatography, eluting with 20% ethyl acetate in hexane, provided the title compound.
1.40.8 methyl 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)amino)acetamido)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxylate The title compound was prepared using the procedure in Example 1.4.7, replacing Example 1.4.6 and Example 1.4.2 with Example 1.40.7 and Example 1.40.5, respectively. MS (ESI) m/e 797.4 (M+H)+.
1.40.9 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)amino)acetamido)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxylic acid
The title compound was prepared using the procedure in Example 1.26.8, replacing Example 1.26.7 with Example 1.40.8. MS (ESI) m/e 783.4 (M+H)+.
1.40.10 tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-((tert- butoxycarbonyl)amino)acetamido)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate
The title compound was prepared using the procedure in Example 1.26.9, replacing Example 1.26.8 with Example 1.40.9. MS (ESI) m/e 915.3 (M+H)+.
1.40.11 3-(1-{[3-(2-aminoacetamido)-5,7-dimethyltricyclo[3.3.1.13,7]decan-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-{8-[(1,3-benzothiazol-2- yl)carbamoyl]-3,4-dihydroisoquinolin-2(1H)-yl}pyridine-2-carboxylic acid
The title compound was prepared using the procedure in Example 1.26.10, replacing Example 1.26.9 with Example 1.40.10. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ 12.82 (s, 1H), 8.00 (dd, 1H), 7.90– 7.79 (m, 4H), 7.76 (d, 1H), 7.59 (dd, 1H), 7.49– 7.38 (m, 3H), 7.37– 7.29 (m, 2H), 7.25 (s, 1H), 6.92 (d, 1H), 4.92 (s, 2H), 3.85 (t, 2H), 3.77 (s, 2H), 3.40 (q, 2H), 2.98 (t, 2H), 2.07 (s, 3H), 1.63 (s, 2H), 1.57– 1.38 (m, 4H), 1.15– 0.93 (m, 6H), 0.80 (s, 6H). MS (ESI) m/e 759.2 (M+H)+.
1.41 Synthesis of 3-[1-({3-[(2-aminoethyl)sulfanyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]-6-[8- (1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]pyridine-2- carboxylic acid (W3.41)
1.41.1 3-bromo-5,7-dimethyladamantane-1-carboxylic acid
To a solution of bromine (18.75 mL) was added iron (10.19 g) at 0 °C, and the mixture was stirred for 30 minutes. 3,5-Dimethyladamantane-1-carboxylic acid (19 g) was added to the above mixture portionwise. The mixture was stirred at room temperature for 36 hours. After adding ice- water (50 mL) and 6N aqueous HCl (100 mL), the mixture was treated with Na2SO3 (100 g dissolved in 500 mL water). The aqueous layer was extracted with dichloromethane (300 mL x 4). The combined organic layers were washed with 1N aqueous HCl (300 mL) and brine, dried over magnesium sulfate, filtered and concentrated to give the title compound, which was used in the next step without additional purification. 1H NMR: (400 MHz, CDCl3) δ ppm 2.23 (s, 2H), 2.01 - 1.74 (m, 4H), 1.61 - 1.47 (m, 6H), 0.93 (s, 6H). LC-MS (ESI) m/e 285.0 (M+H)+.
1.41.2 3-bromo-5,7-dimethyladamantan-1-yl)methanol
To a solution of Example 1.41.1 (10 g) in tetrahydrofuran (20 mL) was added BH3.THF (69.6 mL). The mixture was stirred at room temperature for 16 hours. Upon the completion of the reaction, methanol (20 mL) was added dropwise, and the resulting mixture was stirred for 30 minutes. The mixture was concentrated under reduced pressure. The residue was purified by column
chromatography on silica gel, eluting with petroleum ether/ethyl acetate (from 8/1 to 5/1), to give the title compound. 1H NMR: (400 MHz, CDCl3) δ ppm 3.28 (s, 2H), 1.98 - 1.95 (m, 6H), 1.38 - 1.18 (m, 7H), 0.93 (s, 6H).
1.41.3 1-((3-bromo-5,7-dimethyladamantan-1-yl)methyl)-1H-pyrazole
A mixture of 2-(tributylphosphoranylidene)acetonitrile (919 mg), 1H-pyrazole (259 mg) and Example 1.41.2 (800 mg) in toluene (8 mL) was stirred at 90 °C for 16 hours. The mixture was concentrated, and the residue was diluted with ethyl acetate (50 mL). The mixture was washed with brine, dried over magnesium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography, eluting with petroleum ether/ethyl acetate, to give the title compound. LC-MS (ESI) m/e 325.1 (M+H)+.
1.41.4 3-((1H-pyrazol-1-yl)methyl)-5,7-dimethyladamantane-1-thiol
A mixture of Example 1.41.3 (2.8g) and thiourea (15.82 g) in 33% (w/w) HBr in acetic acid (50 mL) was stirred at 110 °C for 16 hours and concentrated under reduced pressure to give a residue. The residue was dissolved in 20% ethanol in water (v/v: 200 mL), and sodium hydroxide (19.06 g) was added. The resulting solution was stirred at room temperature for 16 hours and concentrated. The residue was dissolved in water (60 mL), and acidified with 6 N aqueous HCl to pH 5– pH 6. The mixture was extracted with ethyl acetate (200 mL x 2). The combined organic layers were washed with brine, dried over MgSO4, filtered and concentrated to give the title compound. MS (ESI) m/e 319.1 (M+H)+.
1.41.5 2-((-3-((1H-pyrazol-1-yl)methyl)-5,7-dimethyladamantan-1- yl)thio)ethanol
To a solution of Example 1.41.4 (3.3g) in ethanol (120 mL) was added sodium ethoxide (2.437 g). The mixture was stirred for 10 minutes, and 2-chloroethanol (1.80 mL) was added dropwise. The mixture was stirred at room temperature for 6 hours and neutralized with 1 N aqueous HCl to pH 7. The mixture was concentrated, and the residue was extracted with ethyl acetate (200 mL x 2). The combined organic layers were washed with brine, dried over MgSO4, filtered and concentrated. The residue was purified by column chromatography on silica gel, eluting with petroleum ether /ethyl acetate from 6/1 to 2/1, to give the title compound. MS (ESI) m/e 321.2 (M+H)+. 1.41.6 2-((-3,5-dimethyl-7-((5-methyl-1H-pyrazol-1-yl)methyl)adamantan-1- yl)thio)ethanol
To a solution of Example 1.41.5 (2.3 g) in tetrahydrofuran (60 mL) was added n-butyllithium (14.35 mL, 2M in hexane) at -20 °C dropwise under nitrogen. The mixture was stirred for 2 hours. Methyl iodide (4.49 mL) was added to the resulting mixture at -20 °C, and the mixture was stirred at - 20 °C for 2 hours. The reaction was quenched by the dropwise addition of saturated aqueous NH4Cl solution at -20 °C. The resulting mixture was stirred for 10 minutes and acidified with 1 N aqueous HCl to pH 5. The mixture was extracted with ethyl acetate twice. The combined organic layers were washed with brine, dried over MgSO4, filtered and concentrated to give the title compound. MS (ESI) m/e 335.3 (M+H)+.
1.41.7 2-((-3-((4-iodo-5-methyl-1H-pyrazol-1-yl)methyl)-5,7- dimethyladamantan-1-yl)thio)ethanol
To a solution of Example 1.41.6 (3.65 g) in N,N-dimethylformamide (90 mL) was added N- iodosuccinimide (3.68 g). The mixture was stirred at room temperature for 16 hours. The reaction was quenched by the addition of ice-water (8 mL) and saturated aqueous NaS2O3solution (8 mL). The mixture was stirred for an additional 10 minutes and extracted with ethyl acetate (30 mL x 2). The combined organic layers were washed with brine, dried over MgSO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with petroleum ether/ethyl acetate (6/1 to 3/1), to give the title compound. MS (ESI) m/e 461.2 (M+H)+.
1.41.8 di-tert-butyl [2-({3-[(4-iodo-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]decan-1-yl}sulfanyl)ethyl]-2- imidodicarbonate
To a cold solution (0 oC bath) of Example 1.41.7 (3 g) in dichloromethane(100 mL) was added triethylamine (1.181 mL) and mesyl chloride (0.559 mL). The mixture was stirred at room temperature for 4 hours, and the reaction was quenched by the addition of ice-water (30 mL). The mixture was stirred for an additional 10 minutes and was extracted with dichloromethane (50 mL x 2). The combined organic layers were washed with brine, dried over MgSO4, filtered and concentrated under reduced pressure. The residue was dissolved in acetonitrile (100 mL) and NH(Boc)2 (1.695 g) and Cs2CO3 (4.24 g) were added. The mixture was stirred at 85 °C for 16 hours, and the reaction was quenched by the addition of water (20 mL). The mixture was stirred for 10 minutes and was extracted with ethyl acetate (40 mL x 2). The combined organic layers were washed with brine, dried over MgSO4, filtered and concentrated. The residue was purified by silica gel chromatography, eluting with petroleum ether/ethyl acetate from 10/1 to 6/1, to give the title compound. MS (ESI) m/e 660.1 (M+H)+.
1.41.9 methyl 2-[5-(1-{[3-({2-[bis(tert-butoxycarbonyl)amino]ethyl}sulfanyl)- 5,7-dimethyltricyclo[3.3.1.13,7]decan-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-(tert-butoxycarbonyl)pyridin-2-yl]-1,2,3,4- tetrahydroisoquinoline-8-carboxylate
The title compound was prepared using the procedure in Example 1.4.7, replacing Example 1.4.6 and Example 1.4.2 with Example 1.40.7 and Example 1.41.8, respectively. LC-MS (ESI) m/e 900.6 (M+H)+.
1.41.10 2-(6-(tert-butoxycarbonyl)-5-(1-((3-((2-((tert- butoxycarbonyl)amino)ethyl)thio)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-1,2,3,4- tetrahydroisoquinoline-8-carboxylic acid
A slurry of lithium hydroxide (553 mg) in water (4.03 mL) and methanol (4 mL) was cooled to 15 °C. A solution of Example 1.41.9 (800 mg) in tetrahydrofuran (3.23 mL) and methanol (4 mL) was added slowly, and the reaction was stirred at room temperature. After 18 hours the reaction was cooled in an ice-bath and 1.8 g of phosphoric acid in water (4 mL) was added. The biphasic mixture was transferred to a separatory funnel and extracted with ethyl acetate to give the title compound. LC-MS (ESI) m/e 786.2 (M+H)+.
1.41.11 tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-((2-((tert- butoxycarbonyl)amino)ethyl)thio)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinate
A 4 mL amber vial containing Example 1.41.10 (699 mg) was charged with ethyl acetate (5 mL) and 1,1'-carbonyldiimidazole (231 mg) and was stirred for 7 hours at room temperature. A solution of benzo[d]thiazol-2-amine (227 mg) and 1,8-diazabicyclo[5.4.0]undec-7-ene (0.228 mL) in acetonitrile (3 mL) was added, and the reaction was heated to 70 °C. After stirring for 18 hours, the reaction was quenched by the addition of 10 mL 1N aqueous HCl and was extracted with ethyl acetate to give the title compound, which was used in the subsequent step without further purification. MS (ESI) m/e 818.2 (M+H)+.
1.41.12 3-[1-({3-[(2-aminoethyl)sulfanyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}methyl)-5-methyl-1H-pyrazol-4-yl]-6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]pyridine-2-carboxylic acid
To a solution of Example 1.41.11 (510 mg) in dichloromethane (10 mL) was added trifluoroacetic acid (10 mL), and the reaction was stirred at room temperature for 30 minutes. The reaction was quenched with aqueous saturated NaHCO3 solution and extracted with dichloromethane. The product was purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 5- 80% acetonitrile in water containing 0.1% trifluoroacetic acid, to give the title compound. 1H NMR (400 MHz, DMSO-d6) δ ppm 12.86 (bs, 1H), 8.03 (d, 1H), 7.76 (m, 2H), 7.62 (d, 1H), 7.39 (m, 6H), 695 (t 1H) 507 (s 1H) 496 (s 1H) 385 (m 4H) 301 (t 2H) 297 (t 2H) 290 (m 2H) 269 (m 2H), 2.11 (s, 3H), 1.54 (s, 2H), 1.36, (m, 4H), 1.17 (m, 4H), 1.08 (m, 2H), 0.84 (s, 6H). MS (ESI) m/e 762.2 (M+H)+.
1.42 Synthesis of 3-(1-{[3-(3-aminopropyl)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]pyridine-2-carboxylic acid (W3.42) 1.42.1 1-((3-allyl-5,7-dimethyladamantan-1-yl)methyl)-1H-pyrazole
To a solution of Example 1.41.3 (0.825 g) in toluene (5 mL) was added N, N'- azoisobutyronitrile (AIBN, 0.419 g) and allyltributylstannane (2.039 mL). The mixture was purged with N2 stream for 15 minutes, heated at 80 °C for 8 hours and concentrated. The residue was purified by silica gel chromatography, eluting with 5% ethyl acetate in petroleum ether, to provide the title compound. MS (ESI) m/e 285.2 (M+H)+.
1.42.2 1-((3-allyl-5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazole To a solution of Example 1.42.1 (200 mg) in tetrahydrofuran (5 mL) at -78 °C under N2 was added n-butyllithium (2.81 mL, 2.5 M in hexane). The mixture was stirred for 2 hours while the temperature increased to -20 °C and was stirred at -20 °C for 1 hour. Iodomethane (0.659 mL) was added, and the resulting mixture was stirred for 0.5 hour at -20 °C. The reaction was quenched with saturated aqueous NH4Cl solution and extracted with ethyl acetate twice. The organic layer was washed with brine to give the title compound. MS (ESI) m/e 299.2 (M+H)+.
1.42.3 3-(3,5-dimethyl-7-((5-methyl-1H-pyrazol-1-yl)methyl)adamantan-1- yl)propan-1-ol
Under a nitrogen atmosphere, a solution of Example 1.42.2 (2.175 g, 7.29 mmol) in anhydrous tetrahydrofuran (42.5 mL) was cooled to 0 °C. BH3·THF (15.30 mL) was added dropwise. The reaction mixture was stirred at room temperature for 2 hours and cooled to 0 °C. To the reaction mixture was added 10 N aqueous NaOH (5.03 mL) dropwise, followed by 30 percent H2O2 (16.52 mL) water solution. The resulting mixture was warmed to room temperature and stirred for 90 minutes. The reaction was quenched with 10 percent aqueous hydrochloric acid (35 mL). The organic layer was separated, and the aqueous layer was extracted with ethyl acetate (2 x 60 mL). The combined organic layers were washed with brine (3 x 60 mL) and cooled in an ice bath. A saturated aqueous solution of sodium sulfite (15 mL) was carefully added and the mixture was stirred for a few minutes. The organic layer was dried over sodium sulfate, filtered, and concentrated in vacuo. The residue was purified by silica gel chromatography, eluting with petroleum ether/ethyl acetate (3:1 to 1:1), to provide the title compound. MS (ESI) m/e 317.3 (M+H)+.
1.42.4 3-(3-((4-iodo-5-methyl-1H-pyrazol-1-yl)methyl)-5,7- dimethyladamantan-1-yl)propan-1-ol
A mixture of Example 1.42.3 (1.19 g) and 1-iodopyrrolidine-2, 5-dione (1.015 g) in N,N- dimethylformamide (7.5 mL) was stirred for 16 hours at room temperature. The reaction was quenched with saturated aqueous Na2SO3 solution The mixture was diluted with ethyl acetate and washed with saturated aqueous Na2SO3, saturated aqueous Na2CO3 solution, water and brine. The organic layer was dried over anhydrous Na2SO4, filtered, and concentrated. The residue was purified by silica gel chromatography, eluting with petroleum ether/ ethyl acetate (3:1 to 1:1), to provide the title compound. MS (ESI) m/e 443.1 (M+H)+.
1.42.5 3-(3-((4-iodo-5-methyl-1H-pyrazol-1-yl)methyl)-5,7- dimethyladamantan-1-yl)propyl methanesulfonate
To a solution of Example 1.42.4 (1.55 g, 3.50 mmol) in dichloromethane (20 mL) at 0 °C were added triethylamine (0.693 mL) and mesyl chloride (0.374 mL) slowly. The mixture was stirred for 3.5 hours at 20 °C and was diluted with dichloromethane. The organic layer was washed with saturated aqueous NH4Cl, saturated aqueous NaHCO3 solution and brine. The organic layer was dried over Na2SO4, filtered and concentrated to provide the title compound. MS (ESI) m/e 521.1 (M+H)+.
1.42.6 di-tert-butyl (3-{3-[(4-iodo-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]decan-1-yl}propyl)-2-imidodicarbonate To a solution of Example 1.42.5 (1.92 g) in acetonitrile (40 mL) at 20 °C were added di-tert- butyl iminodicarbonate (0.962 g) and Cs2CO3 (2.404 g). The mixture was stirred for 16 hours at 80 °C and diluted with ethyl acetate, washed with water and brine. The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by silica gel chromatography, eluting with petroleum ether/ ethyl acetate (10:1), to provide the title compound. MS (ESI) m/e 642.3 (M+H)+.
1.42.7 methyl 2-[5-{1-[(3-{3-[bis(tert-butoxycarbonyl)amino]propyl}-5,7- dimethyltricyclo[3.3.1.13,7]decan-1-yl)methyl]-5-methyl-1H-pyrazol-4- yl}-6-(tert-butoxycarbonyl)pyridin-2-yl]-1,2,3,4- tetrahydroisoquinoline-8-carboxylate
The title compound was prepared using the procedure in Example 1.4.7, replacing Example 1.4.6 and Example 1.4.2 with Example 1.40.7 and Example 1.42.6, respectively. LC-MS (ESI) m/e 882.6 (M+H)+.
1.42.8 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(3-((tert- butoxycarbonyl)amino)propyl)-5,7-dimethyladamantan-1-yl)methyl)- 5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)-1,2,3,4-tetrahydroisoquinoline- 8-carboxylic acid
The title compound was prepared using the procedure in Example 1.41.10 substituting Example 1.42.7 for Example 1.41.9. LC-MS (ESI) m/e 468.5 (M+H)+.
1.42.9 tert-butyl 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(3-((tert- butoxycarbonyl)amino)propyl)-5,7-dimethyladamantan-1-yl)methyl)- 5-methyl-1H-pyrazol-4-yl)picolinate The title compound was prepared using the procedure in Example 1.41.11 substituting Example 1.42.8 for Example 1.41.10.
1.42.10 3-(1-{[3-(3-aminopropyl)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]pyridine-2-carboxylic acid
The title compound was prepared using the procedure in Example 1.41.12 substituting Example 1.42.9 for Example 1.41.11. 1H NMR (500 MHz, DMSO-d6) δ ppm 12.86 (s, 1H), 8.03 (d, 1H), 7.79 (d, 1H), 7.62 (d, 4H), 7.47 (dt, 3H), 7.36 (q, 2H), 7.27 (s, 1H), 6.95 (d, 1H), 4.95 (s, 2H), 3.77 (s, 2H), 3.01 (t, 2H), 2.72 (q, 2H), 2.09 (s, 3H), 1.45 (t, 2H), 1.18– 1.05 (m, 9H), 1.00 (d, 6H), 0.80 (s, 6H). MS (ESI) m/e 468.5 (M+H)+.
1.43 Synthesis of 3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]decan-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-{5-[(1,3-benzothiazol-2- yl)carbamoyl]quinolin-3-yl}pyridine-2-carboxylic acid (W3.43) 1.43.1 methyl 3-bromoquinoline-5-carboxylate
To a solution of 3-bromoquinoline-5-carboxylic acid (2 g) in methanol (30 mL) was added concentrated H2SO4 (5 mL). The solution was stirred at reflux overnight. The mixture was concentrated under reduced pressure. The residue was dissolved in ethyl acetate (300 mL) and washed with aqueous Na2CO3 solution, water and brine. After drying over anhydrous sodium sulfate, filtration and evaporation of the solvent gave the title product. MS (ESI) m/e 266 (M+H)+.
1.43.2 methyl 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)quinoline-5- carboxylate
To a solution of Example 1.43.1 (356 mg) in N,N-dimethylformamide (5 mL) was added [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (55 mg), potassium acetate (197 mg) and bis(pinacolato)diboron (510 mg). The mixture was stirred at 60 oC overnight. The mixture was cooled to room temperature and used in the next reaction without further work up. MS (ESI) m/e 339.2 (M+Na)+.
1.43.3 methyl 3-[5-{1-[(3-{2-[bis(tert-butoxycarbonyl)amino]ethoxy}-5,7- dimethyltricyclo[3.3.1.13,7]decan-1-yl)methyl]-5-methyl-1H-pyrazol-4-yl}-6- (tert-butoxycarbonyl)pyridin-2-yl]quinoline-5-carboxylate
To a solution of Example 1.43.2 (626 mg) in 1,4-dioxane (10 mL) and water (5 mL) was added Example 1.23.3 (1.46 g), bis(triphenylphosphine)palladium(II) dichloride (140 mg), and CsF (911 mg). The mixture was stirred at 120 oC for 30 minutes under microwave conditions (Biotage Initiator). The mixture was diluted with ethyl acetate (200 mL), washed with water and brine, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography, eluting with 20% ethyl acetate in heptane (1 L) to give the title product. MS (ESI) m/e 8803 (M+H)+ 1.43.4 3-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)amino)ethoxy)-5,7-dimethyladamantan-1-yl)methyl)- 5-methyl-1H-pyrazol-4-yl)pyridin-2-yl)quinoline-5-carboxylic acid To a solution of Example 1.43.3 (1.34 g) in tetrahydrofuran (10 mL), methanol (5 mL) and water (5 mL) was added LiOH H2O (120 mg), and the mixture was stirred at room temperature overnight. The mixture was acidified with 2N aqueous HCl, diluted with ethyl acetate (400 mL), washed with water and brine and dried over anhydrous sodium sulfate. Filtration and evaporation of solvent gave the title product. MS (APCI) m/e 766.3 (M+H)+.
1.43.5 3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]decan-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)-6-{5-[(1,3-benzothiazol-2- yl)carbamoyl]quinolin-3-yl}pyridine-2-carboxylic acid
To a solution of Example 1.43.4 (200 mg) in dichloromethane (10 mL) was added benzo[d]thiazol-2-amine (39.2 mg), 1-ethyl-3-[3-(dimethylamino)propyl]-carbodiimide hydrochloride (50 mg) and 4-dimethylaminopyridine (32 mg). The mixture was stirred at room temperature overnight. The reaction mixture was diluted with ethyl acetate (200 mL), washed with water and brine, dried over anhydrous sodium sulfate, filtered and concentrated. The residue was dissolved in dichloromethane and trifluoroacetic acid (10 mL, 1:1), and the reaction was stirred overnight. The mixture was concentrated, and the residue was dissolved in N,N-dimethylformamide (12 mL) and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1% trifluoroacetic acid, to give the title product. MS (ESI) m/e 742.1 (M+H)+.
1.44 Synthesis of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl]-3-{1-[(3,5-dimethyl-7-{2-[(2- sulfoethyl)amino]ethoxy}tricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H- pyrazol-4-yl}pyridine-2-carboxylic acid (W3.44)
1.44.1 tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]-3-[1-({3,5-dimethyl-7-[(2,2,7,7- tetramethyl-10,10-dioxido-3,3-diphenyl-4,9-dioxa-10^6-thia-13-aza-3- silapentadecan-15-yl)oxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl- 1H-pyrazol-4-yl]pyridine-2-carboxylate
To a solution of Example 1.18.18 (500 mg) in N,N-dimethylformamide (8 mL) was added 4- ((tert-butyldiphenylsilyl)oxy)-2,2-dimethylbutyl ethenesulfonate (334 mg). The reaction was stirred at room temperature overnight and methylamine (0.3 mL) was added to quench the reaction. The resulting mixture was stirred for 20 minutes and purified by reverse-phase chromatography using an Analogix system (C18 column), eluting with 50-100% acetonitrile in water containing 0.1% v/v trifluoroacetic acid, to provide the title compound.
1.44.2 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)- yl]-3-{1-[(35-dimethyl-7-{2-[(2- sulfoethyl)amino]ethoxy}tricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl- 1H-pyrazol-4-yl}pyridine-2-carboxylic acid
Example 1.44.1 (200 mg) in dichloromethane (5 mL) was treated with trifluoroacetic acid (2.5 mL) overnight. The reaction mixture was concentrated and purified by reverse phase chromatography (C18 column), eluting with 20-60% acetonitrile in water containing 0.1% v/v trifluoroacetic acid, to provide the title compound. 1H NMR (500 MHz, dimethylsulfoxide-d6) δ ppm 12.86 (s, 1H), 8.32 (s, 2H), 8.02 (d, 1H), 7.78 (d, 1H), 7.60 (d, 1H), 7.51 (d, 1H), 7.40-7.49 (m, 2H), 7.31-7.39 (m, 2H), 7.27 (s, 1H), 6.95 (d, 1H), 4.94 (s, 2H), 3.87 (t, 2H), 3.81 (s, 2H), 3.15-3.25 (m, 2H), 3.03-3.13 (m, 2H), 3.00 (t, 2H), 2.79 (t, 2H), 2.09 (s, 3H), 1.39 (s, 2H), 1.22-1.34 (m, 4H), 0.94-1.18 (m, 6H), 0.85 (s, 6H). MS (ESI) m/e 854.1 (M+H)+. Example 2: Synthesis of Exemplary Synthons
This example provides synthetic methods for exemplary synthons useful more making ADCs. 2.1 Synthesis of N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-L- valyl-N-{4-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4- tetrahydroquinolin-7-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1- yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]phenyl}-N5-carbamoyl-L- ornithinamide (Synthon BS)
Example 1.1.14 (72 mg) and 4-((S)-2-((S)-2-(6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanamido)-3-methylbutanamido)-5-ureidopentanamido)benzyl (4-nitrophenyl) carbonate (91 mg) in N,N-dimethylformamide (3 mL) was cooled in a water-ice bath and N,N- diisopropylethylamine (0.12 mL) was added. The mixture was stirred at 0 oC for 2 hours and acetic acid (0.057 mL) was added. After concentration of the solvents, the residue was purified via HPLC (20-80% acetonitrile in 0.1% TFA/water) to provide the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 9.98 (s, 1H), 8.40 (s, 1H), 8.06 (d, 1H), 8.00 (d, 1H), 7.74-7.89 (m, 4H), 7.59 (d, 2H), 7.46 (s, 2H), 7.37 (t, 1H), 7.18-7.32 (m, 4H), 6.99 (s, 2H), 6.01 (s, 1H), 4.98 (s, 3H), 4.38 (d, 2H), 3.47 (d, 2H), 3.36 (t, 2H), 3.28 (t, 2H), 2.91-3.10 (m, 2H), 2.79-2.91 (m, 4H), 2.19-2.25 (m, 3H), 2.06-2.20 (m, 2H), 1.89-2.02 (m, 3H), 1.53-1.74 (m, 2H), 1.30-1.55 (m, 8H), 1.06-1.29 (m, 10H), 0.91-1.06 (m, 2H), 0.76-0.89 (m, 12H). MS (ESI) m/e 1356.3 (M+H)+.
2.2 Synthesis of N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-L- valyl-N-{4-[({[2-({3-[(4-{6-[4-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydro-2H-1,4-benzoxazin-6-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]phenyl}-N5-carbamoyl-L- ornithinamide (Synthon DK) To a solution of 4-((S)-2-((S)-2-(6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanamido)-3- methylbutanamido)-5-ureidopentanamido)benzyl 4-nitrophenyl carbonate (57 mg) and Example 1.2.2 (57 mg) in N,N-dimethylformamide (6 mL) was added N,N-diisopropylethylamine (0.5 mL). The mixture was stirred overnight. The mixture was concentrated under vacuum and the residue was diluted with methanol (3 mL) and acetic acid (0.3 mL), loaded onto a 300 g reverse-phase column, and eluted with 30-70% acetonitrile in 0.1% aqueous TFA solution to provide the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 9.97 (s, 1H), , 8.73 (d, 1H), 8.07 (d, 1H), 7.90-7.98 (m, 1H), , 7.71-7.87 (m, 4H), 7.54-7.63 (m, 2H), , 7.45 (d, 1H), 7.32-7.42 (m, 2H), 7.17-7.31 (m, 3H), 6.92-7.03 (m, 3H), 5.88-6.08 (m, 1H), 4.97 (s, 3H), 4.29-4.46 (m, 4H), 4.12-4.26 (m, 4H), 3.86 (s, 3H), 3.21-3.41 (m, 8H), 2.78-3.10 (m, 6H), 2.20 (s, 3H), 1.90-2.18 (m, 3H), 0.92-1.77 (m, 24H), 0.75- 0.88 (m, 6 H). MS (ESI) m/e 1360.2 (M+H)+.
2.3 Synthesis of N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-L- valyl-N-{4-[({[2-({3-[(4-{6-[4-(1,3-benzothiazol-2-ylcarbamoyl)-1-methyl- 1,2,3,4-tetrahydroquinoxalin-6-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]phenyl}-N5-carbamoyl-L- ornithinamide (Synthon DQ)
The title compound was prepared by substituting Example 1.3.2 for Example 1.2.2 in Example 2.2. 1H NMR (500 MHz, dimethyl sulfoxide-d6) δ ppm 9.99 (s, 1H), 8.17-8.35 (m, 1H), 8.07 (d, 1H), 7.89 (d, 1H), 7.71-7.84 (m, 4H), 7.55-7.65 (m, 2H), 7.43 (s, 1H), 7.36 (t, 1H), 7.28 (d, 2H), 7.21 (t, 1H), 6.99 (s, 2H), 6.83 (d, 1H), 5.97 (s, 1H), 5.28-5.51 (m, 2H), 4.98 (s, 2H), 4.32-4.44 (m, 1H), 4.19 (dd, 1H), 3.97-4.13 (m, 2H), 3.85 (s, 2H), 3.29 (d, 3H), 3.00 (s, 3H), 2.80-2.98 (m, 4H), 2.18-2.26 (m, 3H), 1.88-2.17 (m, 3H), 0.91-1.73 (m, 23H), 0.74-0.92 (m, 12 H). MS (ESI) m/e 1373.3 (M+H)+.
2.4 Synthesis of 4-[(1E)-3-({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2- ylcarbamoyl)-1,2,3,4-tetrahydroquinolin-7-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)prop-1-en-1-yl]-2-({N-[6-(2,5-dioxo- 2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-beta-alanyl}amino)phenyl beta-D- glucopyranosiduronic acid (Synthon DJ)
2.4.1 (E)-tert-butyldimethyl((3-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)allyl)oxy)silane
To a flask charged with tert-butyldimethyl(prop-2-yn-1-yloxy)silane (5 g) and
dichloromethane (14.7 mL) under nitrogen atmosphere was added dropwise 4,4,5,5-tetramethyl-1,3,2- dioxaborolane (3.94 g). The mixture was stirred at room temperature for one minute then transferred via cannula to a nitrogen-sparged flask containing Cp2ZrClH (chloridobis(η5- cyclopentadienyl)hydridozirconium Schwartz’s Reagent) (379 mg) The resulting reaction mixture was stirred at room temperature for 16 hours. The mixture was carefully quenched with water (15 mL), and then extracted with diethyl ether (3x 30 mL). The combined organic phases were washed with water (15 mL), dried over MgSO4, filtered, concentrated and purified by silica gel
chromatography, eluting with a gradient from 0-8% ethyl acetatate/heptanes to give the title compound. MS (ESI) m/z 316.0 (M+NH4)+.
2.4.2 (2S,3R,4S,5S,6S)-2-(4-bromo-2-nitrophenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
(2R,3R,4S,5S,6S)-2-Bromo-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (5 g) was dissolved in acetonitrile (100 mL). Ag2O (2.92 g) was added to the solution and the reaction was stirred for 5 minutes at room temperature. 4-Bromo-2-nitrophenol (2.74 g) was added and the reaction mixture was stirred at room temperature for 4 hours. The silver salt residue was filtered through diatomaceous earth and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography eluting with a gradient of 10-70% ethyl acetate in heptanes to provide the title compound. MS (ESI+) m/z 550.9 (M+NH4)+.
2.4.3 (2S,3R,4S,5S,6S)-2-(4-((E)-3-((tert- butyldimethylsilyl)oxy)prop-1-en-1-yl)-2-nitrophenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
Example 2.4.2 (1 g), sodium carbonate (0.595 g), tris(dibenzylideneacetone)dipalladium (Pd2(dba)3) (0.086 g), and 1,3,5,7-tetramethyl-6-phenyl-2,4,8-trioxa-6-phosphaadamantane (0.055 g) were combined in a 3-neck 50-mL round bottom flask equipped with a reflux condenser and the system was degassed with nitrogen. Separately, a solution of Example 2.4.1 (0.726 g) in
tetrahydrofuran (15 mL) was degassed with nitrogen for 30 minutes. This latter solution was transferred via cannula into the flask containing the solid reagents, followed by addition of degassed water (3 mL) via syringe. The reaction was heated to 60 °C for two hours. The reaction mixture was partitioned between ethyl acetate (3x 30 mL) and water (30 mL). The combined organic phases were dried (Na2SO4), filtered, and concentrated. The residue was purified by silica gel chromatography, eluting with a gradient from 0-35% ethyl acetate/heptanes to provide the title compound. MS (ESI+) m/z 643.1 (M+NH4)+.
2.4.4 (2S,3R,4S,5S,6S)-2-(2-amino-4-((E)-3-hydroxyprop-1-en-1- yl)phenoxy)-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5- triyl triacetate
A 500-mL three-neck, nitrogen-flushed flask equipped with a pressure-equalizing addition funnel was charged with zinc dust (8.77 g). A degassed solution of Example 2.4.3 (8.39 g) in tetrahydrofuran (67 mL) was added via cannula. The resulting suspension was chilled in an ice bath and then 6N aqueous HCl (22.3 mL) was added dropwise via addition funnel at such a rate that the internal temperature of the reaction did not exceed 35 °C. After the addition was complete, the reaction mixture was stirred for two hours at room temperature and then filtered through a pad of diatomaceous earth, rinsing with water and ethyl acetate. The filtrate was treated with saturated aqueous NaHCO3 solution until the water layer was no longer acidic, and the mixture was filtered to remove the resulting solids. The filtrate was transferred to a separatory funnel and the layers were separated. The aqueous layer was extracted with ethyl acetate (3x 75 mL) and the combined organic layers were washed with water (100 mL), dried over Na2SO4, filtered, and concentrated. The residue was triturated with diethyl ether and the solid was collected by filtration to give the title compound. MS (ESI+) m/z 482.0 (M+H)+.
2.4.5 (9H-fluoren-9-yl)methyl (3-chloro-3-oxopropyl)carbamate To a solution of 3-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)propanoic acid (5.0 g) in dichloromethane (53.5 mL) was added sulfurous dichloride (0.703 mL). The mixture was stirred at 60 °C for one hour. The mixture was cooled and concentrated to provide the title compound which was used in the next step without further purification.
2.4.6 (2S,3R,4S,5S,6S)-2-(2-(3-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)propanamido)-4-((E)-3- hydroxyprop-1-en-1-yl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
Example 2.4.4 (6.78 g) was dissolved in dichloromethane (50 mL) and the solution was chilled to 0 °C in an ice bath. N,N-Diisopropylethylamine (3.64 g) was added, followed by dropwise addition of a solution of Example 2.4.5 (4.88 g) in dichloromethane (50 mL). The reaction was stirred for 16 hours allowing the ice bath to come to room temperature. Saturated aqueous NaHCO3 solution (100 mL) was added and the layers were separated. The aqueous layer was further extracted with dichloromethane (2x 50 mL). The extracts were dried over Na2SO4, filtered, concentrated and then purified by silica gel chromatography, eluting with a gradient of 5-95% ethyl acetate/heptane, to give an inseparable mixture of starting aniline and desired title compound. This mixture was partitioned between 1N aqueous HCl (40 mL) and a 1:1 mixture of diethyl ether and ethyl acetate (40 mL), and then the aqueous phase was further extracted with ethyl acetate (2x 25 mL). The organic phases were combined, washed with water (2x 25 mL), dried over Na2SO4, filtered, and concentrated to give the title compound. MS (ESI+) m/z 774.9 (M+H)+.
2.4.7 (2S,3R,4S,5S,6S)-2-(2-(3-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)propanamido)-4-((E)-3-(((4- nitrophenoxy)carbonyl)oxy)prop-1-en-1-yl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
Example 2.4.6 (3.57 g) was dissolved in dichloromethane (45 mL) and bis(4- nitrophenyl)carbonate (2.80 g) was added, followed by dropwise addition of N,N- diisopropylethylamine (0.896 g). The reaction was stirred at room temperature for two hours. Silica gel (20 g) was then added to the reaction solution and the mixture was concentrated to dryness under reduced pressure keeping the bath temperature at or below 25 °C The silica residue was loaded atop a column and the crude material was purified by silica gel chromatography, eluting with a gradient from 0-100% ethyl acetate-heptane, providing partially purified title compound which was contaminated with nitrophenol. This material was triturated with methyl tert-butyl ether (250 mL) and the resulting slurry was allowed to sit for 1 hour. The title compound was collected by filtration. Three successive crops were collected in a similar fashion to give the title compound. MS (ESI+) m/z 939.8 (M+H)+.
2.4.8 3-(1-((3-(2-(((((E)-3-(3-(3-aminopropanamido)-4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2- yl)oxy)phenyl)allyl)oxy)carbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (1-(benzo[d]thiazol-2-ylcarbamoyl)-1,2,3,4-tetrahydroquinolin- 7-yl)picolinic acid
Example 1.1.14 (31 mg) and Example 2.4.7 (33.3 mg) in N,N-dimethylformamide (3 mL) at 0 oC was added N,N-diisopropylethylamine (25 µL). The mixture was stirred overnight, diluted with ethyl acetate and washed with water and brine. The organic layer was dried over Na2SO4, filtered, and concentrated. The residue was dissolved in methanol (2 mL) and tetrahydrofuran (1 mL), cooled to 0 oC, and 3 M lithium hydroxide aqueous solution (0.35 mL) was added. The mixture was stirred at 0 oC for 4 hours, concentrated and purified by a Gilson HPLC system (C18 column), eluting with 0- 60% acetonitrile in 0.1% TFA/water to provide the title compound.
2.4.9 4-[(1E)-3-({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)- 1,2,3,4-tetrahydroquinolin-7-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)prop-1-en-1-yl]-2-({N-[6- (2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-beta- alanyl}amino)phenyl beta-D-glucopyranosiduronic acid
To a solution of Example 2.4.8 (19 mg) in N,N-dimethylformamide (2.5 mL) at 0 °C was added 2,5-dioxopyrrolidin-1-yl 6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoate (10 mg) and N,N- diisopropylethylamine (11.08 µL). The mixture was stirred at 0 oC for 15 minutes and a few drops of acetic acid were added. The mixture was purified by a Gilson HPLC system (C18 column), eluting with 20-60% acetonitrile in 0.1% TFA/water to provide the title compound. 1H NMR (500 MHz, dimethyl sulfoxide-d6) δ ppm 9.03 (s, 1H), 8.40 (s, 1H), 8.25 (d, 1H), 8.00 (d, 1H), 7.73-7.91 (m, 4H), 7.46 (s, 2H), 7.37 (t, 1H), 7.29 (d, 1H), 7.22 (t, 1H), 7.08-7.13 (m, 1H), 7.04 (d, 1H), 6.98 (s, 2H), 6.56 (d, 1H), 6.10-6.25 (m, 1H), 4.86 (s, 1H), 4.64 (d, 2H), 3.95 (d, 2H), 3.86 (d, 4H), 3.24-3.41 (m, 4H), 2.79-2.96 (m, 6H), 2.54 (t, 2H), 2.21 (s, 3H), 2.03 (t, 2H), 1.90-1.98 (m, 2H), 1.34-1.52 (m, 6H), 120-130 (m 5H) 089-120 (m 8H) 082 (d 6 H) MS (ESI) m/e 13912 (M+H)+ 2.5 Synthesis of 4-[(1E)-3-({[2-({3-[(4-{6-[4-(1,3-benzothiazol-2- ylcarbamoyl)-1-methyl-1,2,3,4-tetrahydroquinoxalin-6-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)prop-1-en-1-yl]-2-({N-[6-(2,5-dioxo- 2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-beta-alanyl}amino)phenyl beta-D- glucopyranosiduronic acid (Synthon DO)
2.5.1 3-(1-((3-(2-((E)-4-(3-(3-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)propanamido)-4- (((2S,3R,4S,5S,6S)-3,4,5-triacetoxy-6- (methoxycarbonyl)tetrahydro-2H-pyran-2-yl)oxy)phenyl)-N- methylbut-3-enamido)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(4-(benzo[d]thiazol-2- ylcarbamoyl)-1-methyl-1,2,3,4-tetrahydroquinoxalin-6- yl)picolinic acid
To a cold (0 °C) solution of Example 2.4.7 (98 mg) and Example 1.3.2 (91 mg) was added N- ethyl-N-isopropylpropan-2-amine (0.054 mL). The reaction was slowly warmed to room temperature and stirred overnight. The reaction was quenched by the addition of water and ethyl acetate. The layers were separated, and the aqueous was extracted with additional ethyl acetate (2x). The combined organics were dried with anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was used in the subsequent step without further purification. MS (ESI) m/e 1576.8 (M+H)+.
2.5.2 3-(1-((3-(2-(((((E)-3-(3-(3-aminopropanamido)-4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2- yl)oxy)phenyl)allyl)oxy)carbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (4-(benzo[d]thiazol-2-ylcarbamoyl)-1-methyl-1,2,3,4- tetrahydroquinoxalin-6-yl)picolinic acid
To a solution of Example 2.5.1 (158 mg) in tetrahydrofuran/methanol/water (2:1:1, 4 mL) was added lithium hydroxide monohydrate (20 mg). The reaction mixture was stirred overnight. The mixture was concentrated under vacuum, acidified with TFA, and dissolved in dimethyl
sulfoxide/methanol (9 mL) and loaded on an HPLC (Gilson system, eluting with 10-85% acetonitrile in 0.1% TFA in water) for purification to give the pure title compound. MS (ESI) m/e 1228.2 (M+NH4)+.
2.5.3 4-[(1E)-3-({[2-({3-[(4-{6-[4-(1,3-benzothiazol-2-ylcarbamoyl)-1- methyl-1234-tetrahydroquinoxalin-6-yl]-2-carboxypyridin-3- yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)prop-1-en-1-yl]-2-({N-[6- (2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-beta- alanyl}amino)phenyl beta-D-glucopyranosiduronic acid
To a solution of Example 2.5.2 (20 mg) and 2,5-dioxopyrrolidin-1-yl 6-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)hexanoate (6.5 mg) in N,N-dimethylformamide (2 mL) was added N,N- diisopropylethylamine (0.054 mL). The reaction was stirred overnight. The reaction mixture was diluted with methanol (2 mL) and acidified with TFA. The mixture was concentrated and purified on HPLC (Gilson system, eluting with 10-85% acetonitrile in 0.1% TFA in water) to give the pure title compound. 1H NMR (500 MHz, dimethyl sulfoxide-d6) δ ppm 9.03 (s, 1H), 8.25 (s, 2H), 7.85-7.95 (m, 2H), 7.72-7.83 (m, 3H), 7.43 (s, 2H), 7.32-7.37 (m, 1H), 7.17-7.25 (m, 1H), 7.08-7.14 (m, 1H), 7.04 (d, 1H), 6.98 (s, 2H), 6.82 (d, 1H), 6.56 (d, 1H), 6.08-6.25 (m, 1H), 4.82-4.92 (m, 1H), 4.64 (d, 3H), 4.00-4.11 (m, 4H), 3.81-3.94 (m, 6H), 3.27-3.50 (m, 17H), 3.00 (s, 3H), 2.83-2.96 (m, 3H), 2.53- 2.59 (m, 2H), 2.20 (s, 3H), 2.03 (t, 2H), 1.37-1.55 (m, 4H), 0.90-1.29 (m, 10H), 0.82 (d, 6 H). MS (ESI) m/e 1406.2 (M+H)+.
2.6 Synthesis of 4-[(1E)-3-({[2-({3-[(4-{6-[4-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydro-2H-1,4-benzoxazin-6-yl]-2-carboxypyridin-3- yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)prop-1-en-1-yl]-2-({N-[6-(2,5-dioxo- 2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-beta-alanyl}amino)phenyl beta-D- glucopyranosiduronic acid (Synthon DP)
2.6.1 3-(1-((3-(2-((E)-4-(3-(3-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)propanamido)-4- (((2S,3R,4S,5S,6S)-3,4,5-triacetoxy-6- (methoxycarbonyl)tetrahydro-2H-pyran-2-yl)oxy)phenyl)-N- methylbut-3-enamido)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(4-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydro-2H-benzo[b][1,4]oxazin-6- yl)picolinic acid
To a cold (0 °C) solution of Example 2.4.7 (98 mg) and Example 1.2.2 (91 mg) was added N- ethyl-N-isopropylpropan-2-amine (0.054 mL). The reaction was slowly warmed to room temperature and was stirred overnight. The reaction was quenched by the addition of water and ethyl acetate. The layers were separated, and the aqueous layer was extracted twice with additional ethyl acetate. The combined organics were dried with anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue was used in the subsequent step without further purification. MS (ESI) m/e 1547.7 (M+H)+.
2.6.2 3-(1-((3-(2-(((((E)-3-(3-(3-aminopropanamido)-4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2- yl)oxy)phenyl)allyl)oxy)carbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (4-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydro-2H- benzo[b][1,4]oxazin-6-yl)picolinic acid
The title compound was prepared by substituting Example 2.6.1 for Example 2.5.1 in Example 2.5.2. MS (ESI) m/e 1200.1 (M+NH4)+.
2.6.3 4-[(1E)-3-({[2-({3-[(4-{6-[4-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydro-2H-1,4-benzoxazin-6-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)prop-1-en-1-yl]-2-({N-[6- (2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-beta- alanyl}amino)phenyl beta-D-glucopyranosiduronic acid
The title compound was prepared by substituting Example 2.6.2 for Example 2.5.2 in Example 2.5.3. 1H NMR (500 MHz, dimethyl sulfoxide-d6) δ ppm 9.04 (s, 1H), , 8.74 (s, 1H), 8.26 (s, 1H), , 7.96 (d, 1H), 7.71-7.92 (m, 4H), 7.35-7.48 (m, 3H), 7.23 (t, 1H), 7.11 (d, 1H), 6.96-7.07 (m, 4H), 6.57 (d, 1H), 6.11-6.24 (m, 1H), 4.81-4.93 (m, 1H), 4.65 (d, 2H), 4.32-4.40 (m, 2H), 4.17 (s, 3H), 3.23-3.51 (m, 14H), 2.83-2.98 (m, 3H), 2.54 (t, 2H), 2.21 (s, 3H), 2.03 (t, 2H), 1.34-1.55 (m, 6H), 0.92-1.31 (m, 13H), 0.82 (d, 6 H). MS (ESI) m/e 1415.2 (M+Na)+.
2.7 Synthesis of 4-[(1E)-3-({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)naphthalen-2-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)prop-1-en-1-yl]-2-({N-[6-(2,5-dioxo- 2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-beta-alanyl}amino)phenyl beta-D- glucopyranosiduronic acid (Synthon HO)
2.7.1 3-(1-((3-(2-(((((E)-3-(3-(3-aminopropanamido)-4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2- yl)oxy)phenyl)allyl)oxy)carbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (8-(benzo[d]thiazol-2-ylcarbamoyl)naphthalen-2-yl)picolinic acid To a cold (0 °C) solution of Example 2.4.7 (22 mg) and Example 1.6.3 (20 mg) was added N- ethyl-N-isopropylpropan-2-amine (0.054 mL). The reaction was slowly warmed to room temperature and stirred overnight. The reaction was quenched by the addition of water and ethyl acetate. The layers were separated, and the aqueous layer was extracted twice with additional ethyl acetate. The combined organics were dried with anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude title compound which was dissolved in tetrahydrofuran/methanol/water (2:1:1, 4 mL). Lithium hydroxide monohydrate (40 mg) was added, and the reaction mixture stirred overnight. The mixture was then concentrated under vacuum, acidified with TFA, dissolved in dimethyl sulfoxide/methanol and purified on an HPLC (Gilson system, eluting with 10-85% acetonitrile in 0.1% TFA in water) to give the title compound.
2.7.2 4-[(1E)-3-({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)naphthalen-2-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)prop-1-en-1-yl]-2-({N-[6- (2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-beta- alanyl}amino)phenyl beta-D-glucopyranosiduronic acid
The title compound was prepared by substituting Example 2.7.1 for Example 2.5.2 in Example 2.5.3. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 13.09 (s, 1H), 9.02 (s, 2H), 8.37 (d, 1H), 8.12-8.29 (m, 4H), 8.06 (s, 1H), 8.02 (d, 1H), 7.93 (d, 1H), 7.76-7.89 (m, 2H), 7.70 (t, 1H), 7.43-7.54 (m, 2H), 7.37 (t, 1H), 7.00-7.13 (m, 2H), 6.98 (s, 2H), 6.56 (d, 1H), 6.08-6.25 (m, 1H), 4.86 (s, 1H), 4.64 (d, 2H), 3.81-3.94 (m, 6H), 3.18-3.51 (m, 12H), 2.78-2.96 (m, 4H), 2.49-2.59 (m, 2H), 2.22 (s, 3H), , 2.03 (t, 2H), 1.33-1.54 (m, 6H), 0.93-1.30 (m, 12H), 0.82 (d, 6 H). MS (ESI) m/e 1408.3 (M+Na)+.
2.8 Synthesis of 4-[(1E)-3-({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.1~3,7~]dec-1- yl}oxy)ethyl](oxetan-3-yl)carbamoyl}oxy)prop-1-en-1-yl]-2-({N-[6-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-beta-alanyl}amino)phenyl beta-D-glucopyranosiduronic acid (Synthon IT)
2.8.1 3-(1-(((3-(2-(((((E)-3-(3-(3-aminopropanamido)-4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)phenyl)allyl)oxy)carbonyl)(oxetan-3- yl)amino)ethoxy)-5,7-dimethyladamantan-1-yl)methyl)-5- methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl)picolinic acid, Trifluoroacetic Acid To a solution of Example 1.16.7 (0.039 g) and Example 2.4.7 (0.048 g) in N,N- dimethylformamide (1 mL) was added N,N-diisopropylethylamine (0.037 mL), and the reaction was stirred at room temperature for 2 days. The reaction was concentrated, the residue was re-dissolved in a mixture of methanol (0.5 mL) and tetrahydrofuran (0.5 mL) and treated with lithium hydroxide monohydrate (0.027 g) in water (0.5 mL), and the solution was stirred at room temperature. After stirring for 1 hour, the reaction was quenched with trifluoroacetic acid (0.066 mL), diluted with N,N- dimethylformamide (1 mL), and purified by HPLC using a Gilson system eluting with 10-60% acetonitrile in water containing 0.1% v/v trifluoroacetic acid. The desired fractions were combined and freeze-dried to provide the title compound.
2.8.2 4-[(1E)-3-({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)- 3,4-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](oxetan-3- yl)carbamoyl}oxy)prop-1-en-1-yl]-2-({N-[6-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)hexanoyl]-beta-alanyl}amino)phenyl beta-D-glucopyranosiduronic acid
To a solution of Example 2.8.1 (0.024 g) and 2,5-dioxopyrrolidin-1-yl 6-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)hexanoate (8.95 mg) in N,N-dimethylformamide (0.5 mL) was added N- ethyl-N-isopropylpropan-2-amine (0.017 mL), and the reaction was stirred at room temperature for 2 hours. The reaction was diluted with N,N-dimethylformamide (1 mL) and water (1 mL) and was purified by HPLC using a Gilson system eluting with 10-60% acetonitrile in water containing 0.1% v/v trifluoroacetic acid. The desired fractions were combined and freeze-dried to provide the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 12.83 (s, 1H), 9.02 (s, 1H), 8.22 (d, 1H), 8.02 (d, 1H), 7.86 (t, 1H), 7.78 (d, 1H), 7.60 (d, 1H), 7.56-7.39 (m, 3H), 7.39-7.30 (m, 2H), 7.27 (s, 1H), 7.14-6.89 (m, 5H), 6.56 (d, 1H), 4.94 (s, 2H), 4.83 (t, 1H), 4.63 (t, 2H), 4.54 (t, 1H), 3.93-3.83 (m, 6H), 3.83-3.75 (m, 4H), 3.33 (dt, 10H), 2.99 (t, 2H), 2.54 (d, 2H), 2.08 (d, 3H), 2.02 (t, 2H), 1.54- 0.72 (m, 26H). MS (ESI) m/e 1433.3 (M+H)+.
2.9 Synthesis of 4-[(1E)-3-({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2- methoxyethyl)carbamoyl}oxy)prop-1-en-1-yl]-2-({N-[6-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)hexanoyl]-beta-alanyl}amino)phenyl beta-D- glucopyranosiduronic acid (Synthon KA)
2.9.1 3-(1-((3-(2-(((((E)-3-(3-(3-aminopropanamido)-4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)phenyl)allyl)oxy)carbonyl)(2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)- yl)picolinic acid
Example 1.12.10 (150 mg) was dissolved in N,N-dimethylformamide (0.5 mL), and Example 2.4.7 (190 mg) and N-ethyl-N-isopropylpropan-2-amine (0.30 mL) was added. The reaction was stirred at room temperature overnight. Additional Example 2.4.7 (70 mg) and N,N- diisopropylethylamine (0.10 mL) were added and the reaction was allowed to stir another day. The reaction was then concentrated and the residue was dissolved in tetrahydrofuran (2 mL) and methanol (2 mL), then 1.94N aqueous lithium hydroxide monohydrate (1.0 mL) was added and the mixture was stirred at room temperature for one hour. Purification by reverse phase chromatography (C18 column), eluting with 10-90% acetonitrile in 0.1% TFA/water, provided the title compound as a trifluoroacetic acid salt. MS (ESI) m/e 1270.4 (M-H)-.
2.9.2 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-(((((E)-3-(4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)-3-(3-(6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanamido)propanamido)phenyl)allyl)oxy)carbonyl)(2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinic acid
Example 2.9.1 (16 mg) was dissolved in N,N-dimethylformamide (0.3 mL), then 2,5- dioxopyrrolidin-1-yl 6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoate (5 mg) and N-ethyl-N- isopropylpropan-2-amine (11 µL) were added. The reaction mixture was stirred for three hours at room temperature, and purification by reverse phase chromatography (C18 column), eluting with 10- 90% acetonitrile in 0.1% TFA/water, provided the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 12.71 (v br s, 1H), 9.03 (s, 1H), 8.25 (s, 1H), 8.01 (d, 1H), 7.87 (br m, 1H), 7.76 (t, 2H), 7.50 (d, 1H), 7.46 (t, 1H), 7.33 (t, 1H), 7.28 (s, 1H), 7.08 (d, 1H), 7.03 (m, 2H), 6.98 (s, 2H), 6.56 (d, 1H), 6.17 (m,1H), 5.00 (s, 2H), 4.86 (br m, 1 H), 4.64 (d, 2H), 3.88 (m, 6H), 3.79 (br m, 2H), 3.43, 3.35 (m, m, total 16H), 3.22 (s, 3H), 2.80 (m, 2H), 2.54 (m, 2H), 2.09 (s, 3H), 2.03 (t, 2H), 1.45 (m, 6H), 1.37 (br m, 2H), 1.28-0.90 (m, 10H), 0.77-0.82 (m, 6H). MS (ESI) m/e 1463.5 (M-H)-.
2.10 Synthesis of 4-[(1E)-3-({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2- methoxyethyl)carbamoyl}oxy)prop-1-en-1-yl]-2-({N-[(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)acetyl]-beta-alanyl}amino)phenyl beta-D- glucopyranosiduronic acid (Synthon KB) Example 2.9.1 (16 mg) was dissolved in N,N-dimethylformamide (0.3 mL), then 2,5- dioxopyrrolidin-1-yl 2-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetate (4 mg) and N-ethyl-N- isopropylpropan-2-amine (11 µL) were added. The reaction mixture was stirred for three hours at room temperature, and purification by reverse phase chromatography (C18 column), eluting with 10- 90% acetonitrile in 0.1% TFA/water, provided the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 9.06 (s, 1H), 8.25 (br m, 2H), 8.01 (d, 1H), 7.76 (t, 2H), 7.49 (d, 1H), 7.47 (t, 1H), 7.33 (t, 1H), 7.28 (s, 1H), 7.11 (d, 1H), 7.08 (s, 2H), 7.03 (m, 2H), 6.56 (d, 1H), 6.17 (m,1H), 5.00 (s, 2H), 4.86 (br m, 1 H), 4.64 (d, 2H), 4.02 (s, 2H), 3.88 (m, 6H), 3.79 (br m, 2H), 3.43, 3.35 (m, m, total 14H), 3.22 (s, 3H), 2.80 (m, 2H), 2.57 (m, 2H), 2.09 (s, 3H), 1.37 (br m, 2H), 1.28-0.90 (m, 10H), 0.77-0.82 (m, 6H). MS (ESI) m/e 1407.4 (M-1)-.
2.11 Synthesis of 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5- methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-3-[2-(2-{[3-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1- yl)propanoyl]amino}ethoxy)ethoxy]phenyl beta-D-glucopyranosiduronic acid (Synthon KT)
2.11.1 (2S,3R,4S,5S,6S)-2-(4-formyl-3-hydroxyphenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
2,4-Dihydroxybenzaldehyde (15 g) and (2S,3R,4S,5S,6S)-2-bromo-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (10 g) were dissolved in acetonitrile followed by the addition of silver carbonate (10 g) and the reaction was heated to 49 °C. After stirring for 4 hours, the reaction was cooled, filtered and concentrated. The crude title compound was suspended in dichloromethane and was filtered through diatomaceous earth and concentrated. The residue was purified by silica gel chromatography, eluting with ethyl acetate/heptane, to provide the title compound.
2.11.2 (2S,3R,4S,5S,6S)-2-(3-hydroxy-4-(hydroxymethyl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
A solution of Example 2.11.1 (16.12 g) in tetrahydrofuran (200 mL) and methanol (200 mL) was cooled to 0 °C and sodium borohydride (1.476 g) was added portionwise. The reaction was stirred for 20 minutes and quenched with a 1:1 mixture of water:aqueous saturated sodium
bicarbonate solution (400 mL). The resulting solids were filtered off and rinsed with ethyl acetate. The phases were separated and the aqueous layer extracted four times with ethyl acetate. The combined organic layers were dried over magnesium sulfate, filtered, and concentrated. The crude title compound was purified via silica gel chromatography eluting with heptane/ethyl acetate to provide the title compound. MS (ESI) m/e 473.9 (M+NH4)+. 2.11.3 (2S,3R,4S,5S,6S)-2-(4-(((tert-butyldimethylsilyl)oxy)methyl)-3- hydroxyphenoxy)-6-(methoxycarbonyl)tetrahydro-2H-pyran- 3,4,5-triyl triacetate
To Example 2.11.2 (7.66 g) and tert-butyldimethylsilyl chloride (2.78 g) in dichloromethane (168 mL) at -5 °C was added imidazole (2.63 g) and the reaction was stirred overnight allowing the internal temperature of the reaction to warm to 12 °C. The reaction mixture was poured into saturated aqueous ammonium chloride and extracted four times with dichloromethane. The combined organics were washed with brine, dried over magnesium sulfate, filtered and concentrated. The crude title compound was purified via silica gel chromatography eluting with heptane/ethyl acetate to provide the title compound. MS (ESI) m/e 593.0 (M+Na)+.
2.11.4 (2S,3R,4S,5S,6S)-2-(3-(2-(2-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)ethoxy)ethoxy)-4-(((tert- butyldimethylsilyl)oxy)methyl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
To Example 2.11.3 (5.03 g) and triphenylphosphine (4.62 g) in toluene (88 mL) was added di- tert-butyl-azodicarboxylate (4.06 g) and the reaction was stirred for 30 minutes. (9H-Fluoren-9- yl)methyl (2-(2-hydroxyethoxy)ethyl)carbamate was added and the reaction was stirred for an additional 1.5 hours. The reaction was loaded directly onto silica gel and was eluted with
heptane/ethyl acetate to provide the title compound.
2.11.5 (2S,3R,4S,5S,6S)-2-(3-(2-(2-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)ethoxy)ethoxy)-4- (hydroxymethyl)phenoxy)-6-(methoxycarbonyl)tetrahydro-2H- pyran-3,4,5-triyl triacetate
Example 2.11.4 (4.29 g) was stirred in a 3:1:1 solution of acetic acid:water:tetrahydrofuran (100 mL) overnight. The reaction was poured into saturated aqueous sodium bicarbonate and extracted with ethyl acetate. The organic layer was dried over magnesium sulfate, filtered and concentrated. The crude title compound was purified via silica gel chromatography, eluting with heptane/ethyl acetate, to provide the title compound.
2.11.6 (2S,3R,4S,5S,6S)-2-(3-(2-(2-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)ethoxy)ethoxy)-4-((((4- nitrophenoxy)carbonyl)oxy)methyl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
To a solution of Example 2.11.5 (0.595 g) and bis(4-nitrophenyl) carbonate (0.492 g) in N,N- dimethylformamide (4 mL) was added N-ethyl-N-isopropylpropan-2-amine (0.212 mL). After 1.5 hours, the reaction was concentrated under high vacuum. The reaction was loaded directly onto silica gel and eluted using heptane/ethyl acetate to provide the title compound. MS (ESI) m/e 922.9 (M+Na)+ 2.11.7 3-(1-((3-(2-((((2-(2-(2-aminoethoxy)ethoxy)-4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)(2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)- yl)picolinic acid
Example 1.12.10 (150 mg) was dissolved in dimethylformamide (0.5 mL). Example 2.11.6 (190 mg) and N,N-diisopropylethylamine (0.30 mL) were added. The reaction was stirred at room temperature overnight. Then more Example 2.11.6 (70 mg) and more N,N-diisopropylethylamine (0.10 mL) were added and the reaction was allowed to stir for another 24 hours. The reaction was then concentrated and the residue was dissolved in tetrahydrofuran (2 mL) and methanol (2 mL), then 1.94N aqueous lithium hydroxide monohydrate (1.0 mL) was added and the mixture stirred at room temperature for one hour. Purification by reverse phase chromatography (C18 column), eluting with 10-90% acetonitrile in 0.1% TFA/water, provided the title compound as a trifluoroacetic acid salt. MS (ESI) m/e 1261.4 (M-H)-.
2.11.8 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-((((4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)-2-(2-(2-(3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)propanamido)ethoxy)ethoxy)benzyl)oxy)carbonyl)(2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinic acid
Example 2.11.7 (19 mg) was dissolved in dimethylformamide (0.3 mL), then 2,5- dioxopyrrolidin-1-yl 3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)propanoate (6 mg) and N-ethyl-N- isopropylpropan-2-amine (13 µL) were added. The reaction was stirred for three hours at room temperature, then purification by reverse phase chromatography (C18 column), eluting with 10-90% acetonitrile in 0.1% TFA/water, provided the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 12.70 (v br s, 1H), 8.00 (m, 2H), 7.76 (t, 2H), 7.50 (d, 1H), 7.46 (t, 1H), 7.34 (t, 1H), 7.28 (s, 1H), 7.19 (d, 1H), 7.00 (m, 2H), 6.97 (s, 2H), 6.66 (d, 1H), 6.60 (dd, 1H), 5.06 (br m, 1H), 5.00 (s, 2H), 4.96 (s, 2H), 4.09 (m, 2H), 3.88 (m, 6H), 3.80 (br m, 3H), 3.71 (m, 2H), 3.59 (t, 2H), 3.44, 3.38 (both m, total 8H), 3.28 (m, 4H), 3.18 (m, 4H), 2.82 (br m, 2H), 2.33 (t, 2H), 2.09 (s, 3H), 1.33 (br m, 2H), 1.28-0.90 (m, 10H), 0.82 (m, 6H). MS (ESI) m/e 1412.4 (M-H)-.
2.12 Synthesis of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl]-3-(1-{[3-(2-{[({(2E)-3-[4-{[(2S,3R,4S,5S,6S)- 6-carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2-yl]oxy}-3-({3-[({[(2E)- 3-(4-{[(2S 3R 4S 5S 6S)-6-carboxy-345-trihydroxytetrahydro-2H-pyran- 2-yl]oxy}-3-[(3-{[3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)propanoyl]amino}propanoyl)amino]phenyl)prop-2-en-1- yl]oxy}carbonyl)amino]propanoyl}amino)phenyl]prop-2-en-1- yl}oxy)carbonyl](2-methoxyethyl)amino}ethoxy)-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H-pyrazol-4- yl)pyridine-2-carboxylic acid (Synthon KU)
2.12.1 3-(1-((3-(2-(((((E)-3-(3-(3-(((((E)-3-(3-(3- aminopropanamido)-4-(((2S,3R,4S,5S,6S)-6-carboxy-3,4,5- trihydroxytetrahydro-2H-pyran-2- yl)oxy)phenyl)allyl)oxy)carbonyl)amino)propanamido)-4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)phenyl)allyl)oxy)carbonyl)(2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)- yl)picolinic acid
The title compound was isolated as a by-product during the synthesis of Example 2.9.1. MS (ESI) m/e 1708.5 (M-H)-.
2.12.2 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-(((((E)-3-(4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)-3-(3-(((((E)-3-(4-(((2S,3R,4S,5S,6S)-6-carboxy- 3,4,5-trihydroxytetrahydro-2H-pyran-2-yl)oxy)-3-(3-(3-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1- yl)propanamido)propanamido)phenyl)allyl)oxy)carbonyl)amin o)propanamido)phenyl)allyl)oxy)carbonyl)(2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinic acid
The title compound was prepared by substituting Example 2.12.1 for Example 2.11.7 in Example 2.11.8. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 8.99 (s, 1H), 8.97 (s, 1H), 8.17 (br s, 2H), 8.00 (br t, 1H), 7.94 (d, 1H), 7.70 (dd, 2H), 7.41 (m, 2H), 7.27 (t, 1H), 7.04 (br d, 2H), 6.97 (d, 2H), 6.93 (m, 2H), 6.89 (s, 2H), 6.52 (d, 1H), 6.49 (d, 1H), 6.11 (m, 2H), 4.93 (s, 2H), 4.80 (m, 2H), 4.56 (m, 4H), 3.83 (m, 7H), 3.72 (br d, 2H), 3.53 (m, 2H), 3.45-3.28 (m, 28H), 3.15 (s, 3H), 2.74 (m, 2H), 2.48 (m, 4H), 2.26 (t, 2H), 2.02 (s, 3H), 1.28 (br d, 2H), 1.17 (m, 4H), 1.02 (m, 4H), 0.89 (m, 2H), 0.2 (m, 6H). MS (ESI-) m/e 1859.5 (M-H)-.
2.13 Synthesis of 4-[({[2-(2-{2-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-5-methoxy-34-dihydroisoquinolin-2(1H)-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2- methoxyethyl)carbamoyl}oxy)methyl]-5-(beta-D- glucopyranuronosyloxy)phenoxy}ethoxy)ethyl]carbamoyl}oxy)methyl]- 3-[2-(2-{[3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)propanoyl]amino}ethoxy)ethoxy]phenyl beta-D-glucopyranosiduronic acid (Synthon KV)
2.13.1 3-(1-((3-(2-((((2-(2-(2-((((2-(2-(2-aminoethoxy)ethoxy)-4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)amino)ethoxy)ethoxy)-4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)(2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)- yl)picolinic acid
The title compound was isolated as a by-product during the synthesis of Example 2.11.7. MS (ESI) m/e 1690.5 (M- H)-.
2.13.2 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-((((4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)-2-(2-(2-((((4-(((2S,3R,4S,5S,6S)-6-carboxy- 3,4,5-trihydroxytetrahydro-2H-pyran-2-yl)oxy)-2-(2-(2-(3-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1- yl)propanamido)ethoxy)ethoxy)benzyl)oxy)carbonyl)amino)eth oxy)ethoxy)benzyl)oxy)carbonyl)(2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinic acid
The title compound was prepared by substituting Example 2.13.1 for Example 2.11.7 in Example 2.11.8. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 8.00 (m, 2H), 7.76 (t, 2H), 7.50 (d, 1H), 7.46 (m, 1H), 7.34 (m, 1H), 7.28 (s, 1H), 7.19 (m, 3H), 6.99 (m, 2H), 6.97 (s, 2H), 6.66 (m, 2H), 6.60 (m, 2H), 5.07 (m, 2H) 5.00 (s, 2H), 4.96 (s, 2H), 4.93 (s, 2H), 4.09 (m, 4H), 3.90 (m, 7H), 3.80 (br d, 4H), 3.71 (m, 4H), 3.59 (t, 2H), 3.48, 3.44, 3.38 (all m, total 14H), 3.28 (m, 7H), 3.16 (m, 7H), 2.81 (br m, 2H), 2.33 (t, 2H), 2.09 (s, 3H), 1.35 (br d, 2H), 1.28-0.90 (m, 10H), 0.82 (m, 6H). MS (ESI) m/e 1842.5 (M- H)-.
2.14 Synthesis of 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5- methoxy-34-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-3-[2-(2-{[(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetyl]amino}ethoxy)ethoxy]phenyl beta-D-glucopyranosiduronic acid (Synthon KW)
The title compound was prepared by substituting 2,5-dioxopyrrolidin-1-yl 2-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)acetate for 2,5-dioxopyrrolidin-1-yl 3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)propanoate in Example 2.11.8. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 12.73 (v br s, 1H), 8.21 (br t, 1H), 8.01 (d, 1H), 7.76 (t, 2H), 7.50 (d, 1H), 7.46 (t, 1H), 7.34 (t, 1H), 7.28 (s, 1H), 7.19 (d, 1H), 7.07 (s, 2H), 6.99 (t, 2H), 6.66 (d, 1H), 6.60 (dd, 1H), 5.06 (br m, 1H), 5.00 (s, 2H), 4.96 (s, 2H), 4.09 (m, 2H), 4.02 (s, 2H), 3.88 (m, 6H), 3.80 (br m, 3H), 3.71 (m, 2H), 3.48 (t, 2H), 3.39 (m, 6H), 3.28, 3.21 (both m, 8H), 2.82 (br m, 2H), 2.09 (s, 3H), 1.33 (br m, 2H), 1.28-0.90 (m, 10H), 0.831 (m, 6H). MS (ESI) m/e 1398.4 (M-H)-.
2.15 Synthesis of 6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4- tetrahydroquinolin-7-yl]-3-{1-[(3-{[34-(2,5-dioxo-2,5-dihydro-1H-pyrrol- 1-yl)-3-methyl-4,32-dioxo-7,10,13,16,19,22,25,28-octaoxa-3,31- diazatetratriacont-1-yl]oxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl)methyl]-5-methyl-1H-pyrazol-4-yl}pyridine-2-carboxylic acid (Synthon DC)
To a mixture of Example 1.1.14 (30 mg) and 2,5-dioxopyrrolidin-1-yl 1-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)-3-oxo-7,10,13,16,19,22,25,28-octaoxa-4-azahentriacontan-31-oate (MAL- dPEG8-NHS-Ester) (40.8 mg) in N,N-dimethylformamide (3 mL) at 0 °C was added N,N- diisopropylethylamine (48 µL). The mixture was stirred at 0 °C for 20 minutes and at room temperature for 10 minutes. Acetic acid (23 µL) was added and the mixture was purified by reverse phase chromatography (C18 column), eluting with 20-60% acetonitrile in 0.1% TFA/water, to provide the title compound. MS (ESI) m/e 1332.5 (M+H)+.
2.16 Synthesis of 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5- cyano-3,4-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl]carbamoyl}oxy)methyl]-3-[2-(2-{[3-(2,5-dioxo-2,5-dihydro- 1H-pyrrol-1-yl)propanoyl]amino}ethoxy)ethoxy]phenyl beta-D- glucopyranosiduronic acid (Synthon KZ)
2.16.1 3-(1-((3-(2-((((2-(2-(2-aminoethoxy)ethoxy)-4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-5-cyano-3,4- dihydroisoquinolin-2(1H)-yl)picolinic acid The title compound was prepared by substituting Example 1.13.12 for Example 1.12.10 in Example 2.11.7. MS (ESI) m/e 1200 (M+H)+, 1198 (M-H)-.
2.16.2 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-cyano- 3,4-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl]carbamoyl}oxy)methyl]-3-[2-(2-{[3-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1- yl)propanoyl]amino}ethoxy)ethoxy]phenyl beta-D- glucopyranosiduronic acid
The title compound was prepared by substituting Example 2.16.1 for Example 2.11.7 in Example 2.11.8. 1H NMR (400MHz, dimethyl sulfoxide-d6) δ ppm 13.06 (bs, 2H), 8.04 (d, 1H), 8.01 (t, 1H), 7.92 (d, 1H), 7.78 (dd, 2H), 7.53 (d, 1H), 7.48 (t, 1H), 7.37 (t, 1H), 7.29 (s, 1H), 7.19 (d, 1H), 7.06 (t, 1H), 7.03 (d, 1H), 6.98 (s, 1H), 6.65 (d, 1H), 6.59 (dd, 1H), 5.07 (d, 1H), 4.98 (s, 1H), 4.92 (1H), 4.09 (m, 2H), 3.96 (t, 2H), 3.90 (d, 2H), 3.80 (s, 2H), 3.70 (m, 6H), 3.60 (m, 6H), 3.43 (t, 2H), 3.39 (t, 2H), 3.33 (t, 1H), 3.28 (dd, 1H), 3.16 (m, 4H), 3.03 (q, 2H), 2.33 (t, 2H), 2.09 (s, 3H), 1.37 (s, 2H), 1.25 (q, 4H), 1.11 (q, 4H), 1.00 (dd, 2H), 0.83 (s, 6H). MS (ESI) m/e 1351 (M+H)+, 1349 (M- H)-.
2.17 Synthesis of 4-[(1E)-3-({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2- methoxyethyl)carbamoyl}oxy)prop-1-en-1-yl]-2-({N-[3-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)propanoyl]-beta-alanyl}amino)phenyl beta-D- glucopyranosiduronic acid (Synthon LW)
The title compound was prepared by substituting Example 2.9.1 for Example 2.11.7 in Example 2.11.8. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 9.03 (s, 1H), 8.25 (br m, 1H), 8.05 (br t, 1H), 8.01 (d, 1H), 7.76 (t, 2H), 7.49 (d, 1H), 7.47 (t, 1H), 7.33 (t, 1H), 7.28 (s, 1H), 7.10 (d, 1H), 7.05 (m, 1H), 7.00 (m, 2H), 6.96 (s, 2H), 6.56 (d, 1H), 6.17 (m,1H), 5.00 (s, 2H), 4.86 (br m, 1 H), 4.64 (d, 2H), 3.88 (m, 6H), 3.79 (br m, 2H), 3.60 (t, 2H), 3.43, 3.35 (m, m, total 14H), 3.22 (s, 3H), 2.80 (m, 2H), 2.53 (m, 2H), 2.33 (t, 2H), 2.09 (s, 3H), 1.37 (br m, 2H), 1.28-0.90 (m, 10H), 0.82, 0.77 (both s, total 6H). MS (ESI-) m/e 1421.5 (M-H)-.
2.18 Synthesis of N-[(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetyl]-3-sulfo-L- alanyl-N-{5-[(1E)-3-({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5- methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)prop-1-en-1-yl]-2-(beta-D- glucopyranuronosyloxy)phenyl}-beta-alaninamide (Synthon LY) 2.18.1 3-(1-((3-(2-(((((E)-3-(3-(3-((R)-2-amino-3- sulfopropanamido)propanamido)-4-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)phenyl)allyl)oxy)carbonyl)(2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)- yl)picolinic acid
To a solution of (R)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3-sulfopropanoic acid (29 mg) and 2-(3H-[1,2,3]triazolo[4,5-b]pyridin-3-yl)-1,1,3,3-tetramethylisouronium
hexafluorophosphate(V) (28 mg) in N,N-dimethylformamide (0.7 mL) was added N,N- diisopropylethylamine (0.013 mL). After stirring for 2 minutes, the reaction was added to a solution of Example 2.9.1 (70 mg) and N-ethyl-N-isopropylpropan-2-amine (0.035 mL) in N,N- dimethylformamide (0.5 mL) at room temperature, and the mixture was stirred for 3 hours.
Diethylamine (0.035 mL) was added to the reaction and stirring was continued for an additional 2 hours. The reaction was diluted with water (1 mL), and purified by prep HPLC using a Gilson system eluting with 10-85% acetonitrile in water containing 0.1% v/v trifluoroacetic acid. The desired fractions were combined and freeze-dried to provide the title compound. MS (ESI) m/e 1421.4 (M- H).
2.18.2 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-(((((E)-3-(4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)-3-(3-((R)-2-(2-(2,5-dioxo-2,5-dihydro-1H- pyrrol-1-yl)acetamido)-3- sulfopropanamido)propanamido)phenyl)allyl)oxy)carbonyl)(2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinic acid
The title compound was prepared by substituting Example 2.18.1 for Example 2.9.1 in Example 2.10. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 9.12 (s, 1H), 8.32 (d, 1H), 8.22 (br m, 1H), 8.01 (d, 1H), 7.97 (br t, 1H), 7.76 (t, 2H), 7.49 (d, 1H), 7.47 (t, 1H), 7.33 (t, 1H), 7.28 (s, 1H), 7.10 (d, 1H), 7.07 (s, 2H), 7.05 (m, 1H), 7.00 (m, 2H), 6.56 (d, 1H), 6.17 (m, 1H), 5.00 (s, 2H), 4.86 (br m, 1 H), 4.64 (d, 2H), 4.32 (m, 1H), 4.07 (s, 2H), 3.88 (m, 6H), 3.79 (br m, 2H), 3.43, 3.35 (m, m, total 14H), 3.22 (s, 3H), 2.80 (m, 4H), 2.53 (m, 2H), 2.09 (s, 3H), 1.37 (br m, 2H), 1.28-0.90 (m, 10H), 0.82, 0.77 (both s, total 6H). MS (ESI-) m/e 1558.4 (M-H)-. 2.19 Synthesis of N-[3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)propanoyl]-3- sulfo-L-alanyl-N-{5-[(1E)-3-({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2- methoxyethyl)carbamoyl}oxy)prop-1-en-1-yl]-2-(beta-D- glucopyranuronosyloxy)phenyl}-beta-alaninamide (Synthon LZ) The title compound was prepared by substituting Example 2.18.1 for Example 2.11.7 in Example 2.11.8. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 9.12 (s, 1H), 8.22 (br m, 1H), 8.07 (br d, 1H), 8.01 (d, 1H), 7.89 (br t, 1H), 7.76 (t, 2H), 7.49 (d, 1H), 7.47 (t, 1H), 7.33 (t, 1H), 7.28 (s, 1H), 7.10 (d, 1H), 7.05 (m, 1H), 7.00 (m, 2H), 6.96 (s, 2H), 6.56 (d, 1H), 6.17 (m, 1H), 5.00 (s, 2H), 4.86 (br m, 1 H), 4.64 (d, 2H), 4.32 (m, 1H), 3.88 (m, 6H), 3.79 (br m, 2H), 3.60 (t, 2H), 3.43, 3.35 (m, m, total 14H), 3.22 (s, 3H), 2.80 (m, 4H), 2.53 (m, 2H), 2.37 (m, 2H), 2.09 (s, 3H), 1.37 (br m, 2H), 1.28-0.90 (m, 10H), 0.82, 0.77 (both s, total 6H). MS (ESI-) m/e 1572.5 (M-H)-.
2.20 Synthesis of N-[(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetyl]-beta- alanyl-N-{5-[(1E)-3-({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5- methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)prop-1-en-1-yl]-2-(beta-D- glucopyranuronosyloxy)phenyl}-beta-alaninamide (Synthon MB) 2.20.1 3-(1-((3-(2-(((((E)-3-(3-(3-(3- aminopropanamido)propanamido)-4-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)phenyl)allyl)oxy)carbonyl)(2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)- yl)picolinic acid
The title compound was prepared by substituting 3-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)propanoic acid for (R)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)- 3-sulfopropanoic acid in Example 2.18.1. MS (ESI-) m/e 1341.5 (M-H)-.
2.20.2 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-(((((E)-3-(4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)-3-(3-(3-(2-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)acetamido)propanamido)propanamido)phenyl)allyl)oxy)car bonyl)(2-methoxyethyl)amino)ethoxy)-57- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)picolinic acid
The title compound was prepared by substituting Example 2.20.1 for Example 2.9.1 in Example 2.10. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 9.06 (s, 1H), 8.25 (br m, 1H), 8.14 (br t 1H), 8.01 (d, 1H), 7.99 (br m, 1H), 7.76 (t, 2H), 7.49 (d, 1H), 7.47 (t, 1H), 7.33 (t, 1H), 7.28 (s, 1H), 7.10 (d, 1H), 7.07 (s, 2H), 7.05 (m, 1H), 7.00 (m, 2H), 6.56 (d, 1H), 6.17 (m,1H), 5.00 (s, 2H), 4.86 (br m, 1 H), 4.64 (d, 2H), 3.99 (s, 2H), 3.88 (m, 6H), 3.79 (br m, 2H), 3.43, 3.35 (m, m, total 14H), 3.25 (m, 2H), 3.22 (s, 3H), 2.80 (m, 2H), 2.55 (m, 2H), 2.23 (t, 2H), 2.09 (s, 3H), 1.37 (br m, 2H), 1.28-0.90 (m, 10H), 0.82, 0.77 (both s, total 6H). MS (ESI-) m/e 1478.5 (M-H)-.
2.21 Synthesis of N-[3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)propanoyl]- beta-alanyl-N-{5-[(1E)-3-({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2- methoxyethyl)carbamoyl}oxy)prop-1-en-1-yl]-2-(beta-D- glucopyranuronosyloxy)phenyl}-beta-alaninamide (Synthon MC) The title compound was prepared by substituting Example 2.20.1 for Example 2.11.7 in Example 2.11.8. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 9.06 (s, 1H), 8.25 (br m, 1H), 8.01 (d, 1H), 7.94 (br m, 2H), 7.76 (t, 2H), 7.49 (d, 1H), 7.47 (t, 1H), 7.33 (t, 1H), 7.28 (s, 1H), 7.10 (d, 1H), 7.05 (m, 1H), 7.00 (m, 2H), 6.97 (s, 2H), 6.56 (d, 1H), 6.17 (m,1H), 5.00 (s, 2H), 4.86 (br m, 1 H), 4.64 (d, 2H), 3.88 (m, 6H), 3.79 (br m, 2H), 3.60 (t, 2H), 3.43, 3.35 (m, m, total 14H), 3.22 (s, 3H), 3.18 (m, 2H), 2.80 (m, 2H), 2.55 (m, 2H), 2.29 (t, 2H), 2.20 (t, 2H), 2.09 (s, 3H), 1.37 (br m, 2H), 1.28-0.90 (m, 10H), 0.82, 0.77 (both s, total 6H). MS (ESI-) m/e 1492.5 (M-H)-.
2.22 Synthesis of 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5- methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-3-{2-[2-({N-[(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetyl]-3-sulfo-L- alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid
(Synthon ME)
2.22.1 3-(1-((3-(2-((((2-(2-(2-((R)-2-amino-3- sulfopropanamido)ethoxy)ethoxy)-4-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(2-methoxyethyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)picolinic acid The title compound was prepared by substituting Example 2.11.7 for Example 2.9.1 in Example 2.18.1. MS (ESI-) m/e 1412.4 (M-H)-.
2.22.2 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-((((4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)-2-(2-(2-((R)-2-(2-(2,5-dioxo-2,5-dihydro-1H- pyrrol-1-yl)acetamido)-3- sulfopropanamido)ethoxy)ethoxy)benzyl)oxy)carbonyl)(2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinic acid
The title compound was prepared by substituting Example 2.22.1 for Example 2.9.1 in Example 2.10. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 8.32 (d, 1H), 8.02 (d, 1H), 7.76 (m, 3H), 7.52 (d, 1H), 7.46 (t, 1H), 7.34 (t, 1H), 7.30 (s, 1H), 7.19 (d, 1H), 7.06 (s, 2H), 7.00 (m, 2H), 6.66 (d, 1H), 6.58 (dd, 1H), 5.06 (br m, 1H), 5.00 (s, 2H), 4.96 (s, 2H), 4.31 (m, 1H), 4.09 (m, 2H), 4.08 (s, 2H), 3.88 (m, 6H), 3.80 (br m, 4H), 3.71 (m, 2H), 3.44, 3.38 (both m, total 8H), 3.28 (m, 4H), 3.18 (m, 4H), 2.82 (br m, 3H), 2.72 (m, 1H), 2.09 (s, 3H), 1.33 (br m, 2H), 1.28-0.90 (m, 10H), 0.84, 0.81 (both s, total 6H). MS (ESI-) m/e 1549.5 (M-H)-.
2.23 Synthesis of 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5- methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-3-{2-[2-({N-[3-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)propanoyl]-3-sulfo-L- alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid
(Synthon MF)
The title compound was prepared by substituting Example 2.22.1 for Example 2.11.7 in Example 2.11.8. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 12.70 (v br s, 1H), 8.06 (d, 1H), 8.02 (d, 1H), 7.76 (m, 3H), 7.52 (d, 1H), 7.46 (t, 1H), 7.34 (t, 1H), 7.30 (s, 1H), 7.19 (d, 1H), 7.00 (m, 2H), 6.95 (s, 2H), 6.66 (d, 1H), 6.58 (dd, 1H), 5.06 (br m, 1H), 5.00 (s, 2H), 4.96 (s, 2H), 4.31 (m, 1H), 4.09 (m, 2H), 3.88 (m, 6H), 3.80 (br m, 4H), 3.71 (m, 2H), 3.59 (t, 2H), 3.44, 3.38 (both m, total 8H), 3.28 (m, 4H), 3.18 (m, 4H), 2.82 (br m, 3H), 2.72 (m, 1H), 2.33 (m, 2H), 2.09 (s, 3H), 1.33 (br m, 2H), 1.28-0.90 (m, 10H), 0.84, 0.81 (both s, total 6H). MS (ESI-) m/e 1563.5 (M-H)-.
2.24 Synthesis of 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5- methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-3-{2-[2-({N-[(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetyl]-beta- alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid
(Synthon MH)
2.24.1 3-(1-((3-(2-((((2-(2-(2-(3- aminopropanamido)ethoxy)ethoxy)-4-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(2-methoxyethyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)picolinic acid
The title compound was prepared by substituting 3-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)propanoic acid for (R)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)- 3-sulfopropanoic acid and Example 2.11.7 for Example 2.9.1 in Example 2.18.1. MS (ESI-) m/e 1332.5 (M-H)-.
2.24.2 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-((((4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)-2-(2-(2-(3-(2-(2,5-dioxo-2,5-dihydro-1H- pyrrol-1- yl)acetamido)propanamido)ethoxy)ethoxy)benzyl)oxy)carbonyl )(2-methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinic acid
The title compound was prepared by substituting Example 2.24.1 for Example 2.9.1 in Example 2.10. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 12.70 (v br s, 1H), 8.14 (t, 1H), 8.02 (d, 1H), 7.92 (t,1H), 7.76 (t, 2H), 7.52 (d, 1H), 7.46 (t, 1H), 7.34 (t, 1H), 7.28 (s, 1H), 7.19 (d, 1H), 7.06 (s, 2H), 7.00 (m, 2H), 6.66 (d, 1H), 6.58 (dd, 1H), 5.06 (br m, 1H), 5.00 (s, 2H), 4.96 (s, 2H), 4.09 (m, 2H), 3.98 (s, 2H), 3.88 (m, 6H), 3.80 (br m, 4H), 3.71 (m, 2H), 3.44, 3.38 (both m, total 8H), 3.28 (m, 4H), 3.18 (m, 6H), 2.82 (br m, 2H), 2.24 (t, 2H), 2.09 (s, 3H), 1.33 (br m, 2H), 1.28-0.90 (m, 10H), 0.84, 0.81 (both s, total 6H). MS (ESI-) m/e 1469.5 (M-H)-.
2.25 Synthesis of 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5- methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-3-{2-[2-({N-[3-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)propanoyl]-beta- alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid
(Synthon MI)
The title compound was prepared by substituting Example 2.24.1 for Example 2.11.7 in Example 2118 1H NMR (400 MHz dimethyl sulfoxide-d6) δ ppm 1270 (v br s 1H) 802 (d 1H) 7.94 (t,1H), 7.88 (t, 1H), 7.76 (t, 2H), 7.52 (d, 1H), 7.46 (t, 1H), 7.34 (t, 1H), 7.28 (s, 1H), 7.19 (d, 1H), 7.00 (m, 2H), 6.95 (s, 2H), 6.66 (d, 1H), 6.58 (dd, 1H), 5.06 (br m, 1H), 5.00 (s, 2H), 4.96 (s, 2H), 4.09 (m, 2H), 3.88 (m, 6H), 3.80 (br m, 4H), 3.71 (m, 2H), 3.59 (t, 2H), 3.44, 3.38 (both m, total 8H), 3.28 (m, 4H), 3.18 (m, 6H), 2.82 (br m, 2H), 2.30 (t, 2H), 2.20 (t, 2H), 2.09 (s, 3H), 1.33 (br m, 2H), 1.28-0.90 (m, 10H), 0.84, 0.81 (both s, total 6H). MS (ESI-) m/e 1483.5 (M-H)-.
2.26 Synthesis of 2-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5- methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-5-{2-[2-({N-[3-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)propanoyl]-3-sulfo-L- alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid
(Synthon NJ)
2.26.1 4-(2-(2-bromoethoxy)ethoxy)-2-hydroxybenzaldehyde A solution of 2,4-dihydroxybenzaldehyde (1.0 g), 1-bromo-2-(2-bromoethoxy)ethane (3.4 g) and potassium carbonate (1.0 g) were stirred together in acetonitrile (30 mL) and heated to 75 °C. After stirring for 2 days, the reaction was cooled, diluted with ethyl acetate (100 mL), washed with water (50 mL) and brine (50 mL), dried over magnesium sulfate, filtered and concentrated.
Purification via silica gel chromatography, eluting using a gradient of 5-30% ethyl acetate/heptane, provided the title compound. MS (ELSD) m/e 290.4 (M+H)+.
2.26.2 4-(2-(2-azidoethoxy)ethoxy)-2-hydroxybenzaldehyde
To a solution of Example 2.26.1 (1.26 g) in N,N-dimethylformamide (10 mL) was added sodium azide (0.43 g) and the reaction was stirred at room temperature overnight. The reaction was diluted with diethyl ether (100 mL), washed with water (50 mL) and brine (50 mL), dried over magnesium sulfate, filtered, and concentrated. Purification via silica gel chromatography, eluting with a gradient of 5- 30% ethyl acetate/heptane, gave the title compound. MS (ELSD) m/e 251.4 (M+H)+.
2.26.3 (2S,3R,4S,5S,6S)-2-(5-(2-(2-azidoethoxy)ethoxy)-2- formylphenoxy)-6-(methoxycarbonyl)tetrahydro-2H-pyran- 3,4,5-triyl triacetate
A solution of Example 2.26.2 (0.84 g), (3R,4S,5S,6S)-2-bromo-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (1.99 g) and silver (I) oxide (1.16 g) were stirred together in acetonitrile (15 mL). After stirring overnight, the reaction was diluted with dichloromethane (20 mL), diatomaceous earth was added and the reaction filtered and concentrated. Purification via silica gel chromatography, eluting with a gradient of 5-75% ethyl acetate/heptane, gave the title compound. 2.26.4 (2S,3R,4S,5S,6S)-2-(5-(2-(2-azidoethoxy)ethoxy)-2- (hydroxymethyl)phenoxy)-6-(methoxycarbonyl)tetrahydro-2H- pyran-3,4,5-triyl triacetate
A solution of Example 2.26.3 (0.695 g) in methanol (5 mL) and tetrahydrofuran (2 mL) was cooled to 0 °C. Sodium borohydride (0.023 g) was added, and the reaction was warmed to room temperature. After stirring for a total of 1 hour, the reaction was poured into a mixture of ethyl acetate (75 mL) and water (25 mL) and saturated aqueous sodium bicarbonate (10 mL) was added. The organic layer was separated, washed with brine (50 mL), dried over magnesium sulfate, filtered, and concentrated. Purification via silica gel chromatography, eluting with a gradient of 5-85% ethyl acetate/heptane, gave the title compound. MS (ELSD) m/e 551.8 (M-H2O)-.
2.26.5 (2S,3R,4S,5S,6S)-2-(5-(2-(2-aminoethoxy)ethoxy)-2- (hydroxymethyl)phenoxy)-6-(methoxycarbonyl)tetrahydro-2H- pyran-3,4,5-triyl triacetate
To Example 2.26.4 (0.465 g) in tetrahydrofuran (20 mL) was added 5% Pd/C (0.1 g) in a 50 mL pressure bottle and the mixture shaken for 16 hours at 30 psi hydrogen. The reaction was then filtered and concentrated to give the title compound which was used without further purification. MS (ELSD) m/e 544.1 (M+H)+.
2.26.6 (2S,3R,4S,5S,6S)-2-(5-(2-(2-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)ethoxy)ethoxy)-2- (hydroxymethyl)phenoxy)-6-(methoxycarbonyl)tetrahydro-2H- pyran-3,4,5-triyl triacetate
A solution of Example 2.26.5 (0.443 g) in dichloromethane (8 mL) was cooled to 0 °C, then N,N-diisopropylethylamine (0.214 mL) and (9H-fluoren-9-yl)methyl carbonochloridate (0.190 g) were added. After 1 hour, the reaction was concentrated and purified via column chromatography, eluting with 5-95% ethyl acetate/heptane, to give the title compound. MS (ELSD) m/e 748.15 (M- OH)-.
2.26.7 (2S,3R,4S,5S,6S)-2-(5-(2-(2-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)ethoxy)ethoxy)-2-((((4- nitrophenoxy)carbonyl)oxy)methyl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
To a solution of Example 2.26.6 (0.444 g) in N,N-dimethylformamide (5 mL) was added N,N-diisopropylethylamine (0.152 mL) and bis(4-nitrophenyl) carbonate (0.353 g) and the reaction was stirred at room temperature. After 5 hours, the reaction was concentrated and the residue was purified via column chromatography, eluting with 5-90% ethyl acetate/heptane, to give the title compound.
2.26.8 3-(1-((3-(2-((((4-(2-(2-aminoethoxy)ethoxy)-2- (((2S 3R 4S 5S 6S)-6-carboxy-345-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)(2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)- yl)picolinic acid
Example 1.12.10 (360 mg) was dissolved in dimethylformamide (2.5 mL). Example 2.26.7 (450 mg) and N,N-diisopropylethylamine (0.35 mL) were added. The reaction was stirred at room temperature overnight. The reaction was then concentrated and the residue dissolved in
tetrahydrofuran (2.5 mL) and methanol (2.5 mL). Aqueous lithium hydroxide monohydrate (1.94N, 2.2 mL) was added, and the mixture was stirred at room temperature for one hour. Purification by reverse phase chromatography (C18 column), eluting with 10-90% acetonitrile in 0.1% TFA/water, provided the title compound as a trifluoroacetic acid salt. MS (ESI) m/e 1261.4 (M-H)-.
2.26.9 3-(1-((3-(2-((((4-(2-(2-((R)-2-amino-3- sulfopropanamido)ethoxy)ethoxy)-2-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(2-methoxyethyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)picolinic acid
The title compound was prepared by substituting Example 2.26.8 for Example 2.9.1 in Example 2.18.1. MS (ESI-) m/e 1412.4 (M-H)-.
2.26.10 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-((((2- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)-4-(2-(2-((R)-2-(3-(2,5-dioxo-2,5-dihydro-1H- pyrrol-1-yl)propanamido)-3- sulfopropanamido)ethoxy)ethoxy)benzyl)oxy)carbonyl)(2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinic acid
The title compound was prepared by substituting Example 2.26.9 for Example 2.11.7 in Example 2.11.8. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 12.70 (v br s, 1H), 8.06 (d, 1H), 8.02 (d, 1H), 7.76 (t, 3H), 7.52 (d, 1H), 7.46 (t, 1H), 7.34 (t, 1H), 7.30 (s, 1H), 7.19 (d, 1H), 7.00 (m, 2H), 6.95 (s, 2H), 6.70 (d, 1H), 6.58 (dd, 1H), 5.06 (br m, 1H), 5.00 (s, 2H), 4.96 (s, 2H), 4.31 (m, 1H), 4.09 (m, 2H), 3.88 (m, 6H), 3.80 (br m, 4H), 3.71 (m, 2H), 3.59 (t, 2H), 3.44, 3.38 (both m, total 8H), 3.28 (m, 4H), 3.18 (m, 4H), 2.82 (br m, 3H), 2.72 (m, 1H), 2.33 (m, 2H), 2.09 (s, 3H), 1.33 (br m, 2H), 1.28-0.90 (m, 10H), 0.84, 0.81 (both s, total 6H). MS (ESI-) m/e 1563.5 (M-H)-. 2.27 Synthesis of 2-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5- methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-5-{2-[2-({N-[6-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L- alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid
(Synthon NK)
The title compound was prepared by substituting Example 2.26.9 for Example 2.9.1 in Example 2.9.2. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 12.70 (v br s, 1H), 8.06 (d, 1H), 8.02 (d, 1H), 7.76 (t, 3H), 7.52 (d, 1H), 7.46 (t, 1H), 7.34 (t, 1H), 7.30 (s, 1H), 7.19 (d, 1H), 7.00 (m, 2H), 6.95 (s, 2H), 6.70 (d, 1H), 6.58 (dd, 1H), 5.06 (br m, 1H), 5.00 (s, 2H), 4.96 (s, 2H), 4.31 (m, 1H), 4.09 (m, 2H), 3.88 (m, 6H), 3.80 (br m, 4H), 3.71 (m, 2H), 3.59 (t, 2H), 3.44, 3.38 (both m, total 8H), 3.28 (m, 4H), 3.18 (m, 4H), 2.82 (br m, 3H), 2.72 (m, 1H), 2.33 (m, 2H), 2.09 (s, 3H), 1.46 (br m, 4H) 1.33 (br m, 2H), 1.28-0.90 (m, 12H), 0.84, 0.81 (both s, total 6H). MS (ESI-) m/e 1605.4 (M- H)-.
2.28 Synthesis of 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5- methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-3-[3-({N-[6-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L- alanyl}amino)propoxy]phenyl beta-D-glucopyranosiduronic acid (Synthon NL)
2.28.1 (2S,3R,4S,5S,6S)-2-(3-(3-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)propoxy)-4-formylphenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
To a solution of (9H-fluoren-9-yl)methyl (3-hydroxypropyl)carbamate (0.245 g) and triphenylphosphine (0.216 g) in tetrahydrofuran (2 mL) at 0 °C was added diisopropyl
azodicarboxylate (0.160 mL) dropwise. After stirring for 15 minutes, Example 2.11.1 (0.250 g) was added, the ice bath was removed, and the reaction was allowed to warm to room temperature. After 2 hours, the reaction was concentrated, loaded onto silica gel, and eluted using a gradient of 5-70% ethyl acetate/hexanes to give the title compound. MS (APCI) m/e 512.0 (M-FMOC)-.
2.28.2 (2S,3R,4S,5S,6S)-2-(3-(3-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)propoxy)-4- (hydroxymethyl)phenoxy)-6-(methoxycarbonyl)tetrahydro-2H- pyran-3,4,5-triyl triacetate
To a suspension of Example 2.28.1 (0.233 g) in methanol (3 mL) and tetrahydrofuran (1 mL) was added sodium borohydride (6 mg) After 30 minutes the reaction was poured into ethyl acetate (50 mL) and water (25 mL), followed by the addition of sodium bicarbonate (5 mL). The organic layer was separated, washed with brine (25 mL), dried over magnesium sulfate, filtered, and concentrated. Silica gel chromatography, eluting with a gradient of 5- 80% ethyl acetate/heptane, gave the title compound. MS (APCI) m/e 718.1 (M-OH)-.
2.28.3 (2S,3R,4S,5S,6S)-2-(3-(3-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)propoxy)-4-((((4- nitrophenoxy)carbonyl)oxy)methyl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
To a solution of Example 2.28.2 (0.140 g) and bis(4-nitrophenyl) carbonate (0.116 g) in N,N- dimethylformamide (1 mL) was added N-ethyl-N-isopropylpropan-2-amine (0.050 mL). After 1.5 hours, the reaction was concentrated under high vacuum, loaded onto silica gel, and eluted using a gradient of 10-70% ethyl acetate/heptane to give the title compound.
2.28.4 3-(1-((3-(2-((((2-(3-aminopropoxy)-4-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(2-methoxyethyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)picolinic acid
The title compound was prepared by substituting Example 2.28.3 for Example 2.26.7 in Example 2.26.8. MS (ESI-) m/e 1231.3 (M-H)-.
2.28.5 3-(1-((3-(2-((((2-(3-((R)-2-amino-3- sulfopropanamido)propoxy)-4-(((2S,3R,4S,5S,6S)-6-carboxy- 3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(2-methoxyethyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)picolinic acid
The title compound was prepared by substituting Example 2.28.4 for Example 2.9.1 in Example 2.18.1. MS (ESI-) m/e 1382.4 (M-H)-.
2.28.6 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-((((4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)-2-(3-((R)-2-(6-(2,5-dioxo-2,5-dihydro-1H- pyrrol-1-yl)hexanamido)-3- sulfopropanamido)propoxy)benzyl)oxy)carbonyl)(2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinic acid The title compound was prepared by substituting Example 2.28.5 for Example 2.9.1 in Example 2.9.2. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 8.01 (d, 1H), 7.85 (m, 2H), 7.76 (m, 2H), 7.52 (d, 1H), 7.46 (t, 1H), 7.34 (m, 1H), 7.30 (s, 1H), 7.16 (d, 1H), 7.00 (m, 3H), 6.97 (s, 2H), 6.64 (d, 1H), 6.56 (dd, 1H), 5.04 (br m, 1H), 5.00 (s, 2H), 4.96 (s, 2H), 4.28 (m, 1H), 3.97 (m, 2H), 3.88 (m, 6H), 3.80 (m, 2H), 3.71 (m, 2H), 3.37 (m, 8H), 3.27 (m, 4H), 3.17 (m, 4H), 2.90-2.65 (m, 4H), 2.09 (s, 3H), 2.05 (t, 2H), 1.81 (m, 2H), 1.46 (br m, 4H), 1.33 (br m, 2H), 1.28-0.90 (m, 12H), 0.84, 0.81 (both s, total 6H). MS (ESI-) m/e 1575.5 (M-H)-.
2.29 Synthesis of 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5- methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-[3-({N-[6-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L- alanyl}amino)propoxy]phenyl beta-D-glucopyranosiduronic acid (Synthon NM)
2.29.1 3-(1-((3-(2-((((2-(3-aminopropoxy)-4-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)picolinic acid
The title compound was prepared by substituting Example 2.28.3 for Example 2.26.7 and Example 1.9.11 for Example 1.12.10 in Example 2.26.8. MS (ESI-) m/e 1187.4 (M-H)-.
2.29.2 3-(1-((3-(2-((((2-(3-((R)-2-amino-3- sulfopropanamido)propoxy)-4-(((2S,3R,4S,5S,6S)-6-carboxy- 3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)picolinic acid
The title compound was prepared by substituting Example 2.29.1 for Example 2.9.1 in Example 2.18.1. MS (ESI-) m/e 1338.3 (M-H)-.
2.29.3 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-((((4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)-2-(3-((R)-2-(6-(2,5-dioxo-2,5-dihydro-1H- pyrrol-1-yl)hexanamido)-3- sulfopropanamido)propoxy)benzyl)oxy)carbonyl)(methyl)amin o)ethoxy)-5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H- pyrazol-4-yl)picolinic acid
The title compound was prepared by substituting Example 2.29.2 for Example 2.9.1 in Example 2.9.2. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 8.01 (d, 1H), 7.85 (m, 2H), 7.76 (m, 2H), 7.52 (d, 1H), 7.46 (t, 1H), 7.34 (m, 1H), 7.30 (s, 1H), 7.16 (d, 1H), 7.00 (m, 3H), 6.97 (s, 2H), 6.64 (d, 1H), 6.56 (dd, 1H), 5.04 (br m, 1H), 5.00 (s, 2H), 4.96 (s, 2H), 4.28 (m, 1H), 3.97 (m, 2H), 3.88 (m, 6H), 3.80 (m, 2H), 3.44 (m, 6H), 3.28 (m, 4H), 3.17 (m, 2H), 2.90-2.65 (m, 4H), 2.09 (s, 3H), 2.05 (t, 2H), 1.81 (m, 2H), 1.46 (br m, 4H), 1.33 (br m, 2H), 1.28-0.90 (m, 12H), 0.84, 0.81 (both s, total 6H). MS (ESI-) m/e 1531.5 (M-H)-.
2.30 Synthesis of N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-L- valyl-N-{4-[({[(3S)-1-{8-(1,3-benzothiazol-2-ylcarbamoyl)-2-[6-carboxy- 5-(1-{[3-(2-methoxyethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)pyridin-2-yl]-1,2,3,4- tetrahydroisoquinolin-6-yl}pyrrolidin-3- yl]carbamoyl}oxy)methyl]phenyl}-L-alaninamide (Synthon NR) Example 1.17.10 (40 mg) was dissolved in dimethyl sulfoxide (0.3 mL), and 4-((S)-2-((S)-2- (6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanamido)-3-methylbutanamido)propanamido)benzyl (4- nitrophenyl) carbonate (31 mg) and triethylamine (33 µL) were added. The reaction mixture was stirred for 72 hours at room temperature, and purification by reverse phase chromatography (C18 column), eluting with 10-90% acetonitrile in 0.1% TFA water, provided the title compound. MS (ESI) m/e 1357.4 (M+H)+, 1355.5 (M-H)-.
2.31 Synthesis of N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-L- valyl-N-{4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-sulfamoylethyl)carbamoyl}oxy)methyl]phenyl}-N5- carbamoyl-L-ornithinamide (Synthon EB)
The title compound was prepared as described in previous examples. 1H NMR (500 MHz, dimethyl sulfoxide-d6) δ ppm 12.85 (s, 1H), 9.98 (s, 1H), 8.00-8.09 (m, 2H), 7.78 (t, 2H), 7.61 (t, 3H), 7.40-7.53 (m, 3H), 7.33-7.39 (m, 2H), 7.25-7.30 (m, 3H), 6.86-7.00 (m, 5H), 5.99 (s, 1H), 4.86-5.10 (m, 4H), 4.38 (s, 1H), 4.10-4.26 (m, 1H), 3.88 (t, 2H), 3.80 (d, 2H), 3.33-3.39 (m, 2H), 3.30 (d, 2H), 3.18-3.26 (m, 2H), 2.88-3.06 (m, 5H), 2.04-2.24 (m, 5H), 1.87-2.00 (m, 1H), 1.28-1.74 (m, 10H), 0.89-1.27 (m, 12H), 0.74-0.87 (m, 12H). MS (ESI) m/e 1451.3 (M+H)+.
2.32 Synthesis of Control Synthon 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-2-({N-[6-(25-dioxo-25- dihydro-1H-pyrrol-1-yl)hexanoyl]-beta-alanyl}amino)phenyl beta-D- glucopyranosiduronic acid (Synthon H)
2.32.1 (2S,3R,4S,5S,6S)-2-(4-formyl-2-nitrophenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
To a solution of (2R,3R,4S,5S,6S)-2-bromo-6-(methoxycarbonyl)tetrahydro-2H- pyran-3,4,5- triyl triacetate (4 g) in acetonitrile (100 mL)) was added silver(I) oxide (10.04 g) and 4-hydroxy-3- nitrobenzaldehyde (1.683 g). The reaction mixture was stirred for 4 hours at room temperature and filtered. The filtrate was concentrated, and the residue was purified by silica gel chromatography, eluting with 5-50% ethyl acetate in heptanes, to provide the title compound. MS (ESI) m/e (M+18)+.
2.32.2 (2S,3R,4S,5S,6S)-2-(4-(hydroxymethyl)-2-nitrophenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
To a solution of Example 2.32.1 (6 g) in a mixture of chloroform (75 mL) and isopropanol (18.75 mL) was added 0.87 g of silica gel. The resulting mixture was cooled to 0 °C, NaBH4 (0.470 g) was added, and the resulting suspension was stirred at 0 °C for 45 minutes. The reaction mixture was diluted with dichloromethane (100 mL) and filtered through diatomaceous earth. The filtrate was washed with water and brine and concentrated to give the crude product, which was used without further purification. MS (ESI) m/e (M+NH4)+:
2.32.3 (2S,3R,4S,5S,6S)-2-(2-amino-4-(hydroxymethyl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
A stirred solution of Example 2.32.2 (7 g) in ethyl acetate (81 mL) was hydrogenated at 20 °C under 1 atmosphere H2, using 10% Pd/C (1.535 g) as a catalyst for 12 hours. The reaction mixture was filtered through diatomaceous earth, and the solvent was evaporated under reduced pressure. The residue was purified by silica gel chromatography, eluting with 95/5 dichloromethane/methanol, to give the title compound.
2.32.4 3-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)propanoic acid
3-Aminopropanoic acid (4.99 g) was dissolved in 10% aqueous Na2CO3 solution (120 mL) in a 500 mL flask and cooled with an ice bath. To the resulting solution, (9H-fluoren-9-yl)methyl carbonochloridate (14.5 g) in 1,4-dioxane (100 mL) was gradually added. The reaction mixture was stirred at room temperature for 4 hours, and water (800 mL) was then added. The aqueous phase layer was separated from the reaction mixture and washed with diethyl ether (3 x 750 mL). The aqueous layer was acidified with 2N HCl aqueous solution to a pH value of 2 and extracted with ethyl acetate (3 x 750 mL). The organic layers were combined and concentrated to obtain crude product. The crude product was recrystallized in a mixed solvent of ethyl acetate: hexane 1:2 (300 mL) to give the title compound.
2.32.5 (9H-fluoren-9-yl)methyl (3-chloro-3-oxopropyl)carbamate To a solution of Example 2.32.4 in dichloromethane(160 mL) was added sulfurous dichloride (50 mL). The mixture was stirred at 60 °C for 1 hour. The mixture was cooled and concentrated to give the title compound.
2.32.6 (2S,3R,4S,5S,6S)-2-(2-(3-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)propanamido)-4- (hydroxymethyl)phenoxy)-6-(methoxycarbonyl)tetrahydro-2H- pyran-3,4,5-triyl triacetate
To a solution of Example 2.32.3 (6 g) in dichloromethane (480 mL) was added N,N- diisopropylethylamine (4.60 mL). Example 2.32.5 (5.34 g) was added, and the mixture was stirred at room temperature for 30 minutes. The mixture was poured into saturated aqueous sodium bicarbonate and was extracted with ethyl acetate. The combined extracts were washed with water and brine and were dried over sodium sulfate. Filtration and concentration gave a residue that was purified via radial chromatography, using 0-100% ethyl acetate in petroleum ether as mobile phase, to give the title compound.
2.32.7 (2S,3R,4S,5S,6S)-2-(2-(3-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)propanamido)-4-((((4- nitrophenoxy)carbonyl)oxy)methyl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
To a mixture of Example 2.32.6 (5.1 g) in N,N-dimethylformamide (200 mL) was added bis(4-nitrophenyl) carbonate (4.14 g) and N,N-diisopropylethylamine (1.784 mL). The mixture was stirred for 16 hours at room temperature and concentrated under reduced pressure. The crude material was dissolved in dichloromethane and aspirated directly onto a 1 mm radial Chromatotron plate and eluted with 50-100% ethyl acetate in hexanes to give the title compound. MS (ESI) m/e (M+H)+.
2.32.8 3-bromo-5,7-dimethyladamantanecarboxylic acid
In a 50 mL round-bottomed flask at 0°C was added bromine (16 mL). Iron powder (7 g) was then added, and the reaction was stirred at 0°C for 30 minutes. 3,5-Dimethyladamantane-1-carboxylic acid (12 g) was then added. The mixture was warmed up to room temperature and stirred for 3 days. A mixture of ice and concentrated HCl was poured into the reaction mixture. The resulting suspension was treated twice with Na2SO3 (50 g in 200 mL water) to destroy bromine and was extracted three times with dichloromethane. The combined organics were washed with 1N aqueous HCl, dried over Na2SO4, filtered, and concentrated to give the crude title compound.
2.32.9 3-bromo-5,7-dimethyladamantanemethanol
To a solution of Example 2.32.8 (15.4 g) in tetrahydrofuran (200 mL) was added BH3 (1M in tetrahydrofuran, 150 mL). The mixture was stirred at room temperature overnight. The reaction mixture was then carefully quenched by adding methanol dropwise. The mixture was then concentrated under vacuum, and the residue was balanced between ethyl acetate (500 mL) and 2N aqueous HCl (100 mL) The aqueous layer was further extracted twice with ethyl acetate and the combined organic extracts were washed with water and brine, dried over Na2SO4, and filtered.
Evaporation of the solvent gave the title compound.
2.32.10 1-((3-bromo-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl)- 1H-pyrazole
To a solution of Example 2.32.9 (8.0 g) in toluene (60 mL) was added 1H-pyrazole (1.55 g) and cyanomethylenetributylphosphorane (2.0 g). The mixture was stirred at 90oC overnight. The reaction mixture was then concentrated and the residue was purified by silica gel column
chromatography (10:1 heptane:ethyl acetate) to give the title compound. MS (ESI) m/e 324.2 (M+H)+.
2.32.11 2-{[3,5-dimethyl-7-(1H-pyrazol-1- ylmethyl)tricyclo[3.3.1.13,7]dec-1-yl]oxy}ethanol
To a solution of Example 2.32.10 (4.0 g) in ethane-1,2-diol (12 mL) was added triethylamine (3 mL). The mixture was stirred at 150oC under microwave conditions (Biotage Initiator) for 45 minutes. The mixture was poured into water (100 mL) and extracted three times with ethyl acetate. The combined organic extracts were washed with water and brine, dried over Na2SO4, and filtered. Evaporation of the solvent gave the crude product, which was purified by silica gel chromatography, eluting with 20% ethyl acetate in heptane, followed by 5% methanol in dichloromethane, to give the title compound. MS (ESI) m/e 305.2 (M+H)+.
2.32.12 2-({3,5-dimethyl-7-[(5-methyl-1H-pyrazol-1- yl)methyl]tricyclo[3.3.1.13,7]dec-1-yl}oxy)ethanol
To a cooled (-78oC) solution of Example 2.32.11 (6.05 g) in tetrahydrofuran (100 mL) was added n-BuLi (40 mL, 2.5M in hexane). The mixture was stirred at -78oC for 1.5 hours. Iodomethane (10 mL) was added through a syringe, and the mixture was stirred at -78oC for 3 hours. The reaction mixture was then quenched with aqueous NH4Cl and extracted twice with ethyl acetate, and the combined organic extracts were washed with water and brine. After drying over Na2SO4, the solution was filtered and concentrated, and the residue was purified by silica gel column chromatography, eluting with 5% methanol in dichloromethane, to give the title compound. MS (ESI) m/e 319.5 (M+H)+.
2.32.13 1-({3,5-dimethyl-7-[2-(hydroxy)ethoxy]tricyclo[3.3.1.13,7]dec- 1-yl}methyl)-4-iodo-5-methyl-1H-pyrazole
To a solution of Example 2.32.12 (3.5 g) in N,N-dimethylformamide (30 mL) was added N- iodosuccinimide (3.2 g). The mixture was stirred at room temperature for 1.5 hours. The reaction mixture was then diluted with ethyl acetate (600 mL) and washed with aqueous NaHSO3, water, and brine. After drying over Na2SO4, the solution was filtered and concentrated and the residue was purified by silica gel chromatography (20% ethyl acetate in dichloromethane) to give the title compound. MS (ESI) m/e 445.3 (M+H)+. 2.32.14 2-({3-[(4-iodo-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl
methanesulfonate
To a cooled solution of Example 2.32.13 (6.16 g) in dichloromethane (100 mL) was added triethylamine (4.21 g) followed by methanesulfonyl chloride (1.6 g). The mixture was stirred at room temperature for 1.5 hours. The reaction mixture was then diluted with ethyl acetate (600 mL) and washed with water and brine. After drying over Na2SO4, the solution was filtered and concentrated, and the residue was used in the next reaction without further purification. MS (ESI) m/e 523.4 (M+H)+ .
2.32.15 1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-4- iodo-5-methyl-1H-pyrazole
A solution of Example 2.32.14 (2.5 g) in 2M methylamine in methanol (15 mL) was stirred at 100oC for 20 minutes under microwave conditions (Biotage Initiator). The reaction mixture was concentrated under vacuum. The residue was then diluted with ethyl acetate (400 mL) and washed with aqueous NaHCO3, water and brine. After drying over Na2SO4, the solution was filtered and concentrated, and the residue was used in the next reaction without further purification. MS (ESI) m/e 458.4 (M+H)+.
2.32.16 tert-butyl [2-({3-[(4-iodo-5-methyl-1H-pyrazol-1-yl)methyl]- 5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl]methylcarbamate
To a solution of Example 2.32.15 (2.2 g) in tetrahydrofuran (30 mL) was added di-tert-butyl dicarbonate (1.26 g) and a catalytic amount of 4-dimethylaminopyridine. The mixture was stirred at room temperature for 1.5 hours and diluted with ethyl acetate (300 mL). The solution was washed with saturated aqueous NaHCO3, water (60 mL), and brine (60 mL). The organic layer was dried with Na2SO4, filtered, and concentrated. The residue was purified by silica gel chromatography, eluting with 20% ethyl acetate in dichloromethane, to give the title compound. MS (ESI) m/e 558.5 (M+H)+.
2.32.17 6-fluoro-3-bromopicolinic acid
A slurry of 6-amino-3-bromopicolinic acid (25 g) in 400 mL 1:1 dichloromethane/chloroform was added to nitrosonium tetrafluoroborate (18.2 g) in dichloromethane (100 mL) at 5oC over 1 hour, and the resulting mixture was stirred for another 30 minutes, then warmed to 35oC and stirred overnight. The reaction was cooled to room temperature, and then adjusted to pH 4 with aqueous NaH2PO4 solution. The resulting solution was extracted three times with dichloromethane, and the combined extracts were washed with brine, dried over sodium sulfate, filtered and concentrated to provide the title compound.
2.32.18 Tert-butyl 3-bromo-6-fluoropicolinate Para-toluenesulfonyl chloride (27.6 g) was added to a solution of Example 2.32.17 (14.5 g) and pyridine (26.7 mL) in dichloromethane (100 mL) and tert-butanol (80 mL) at 0oC. The reaction was stirred for 15 minutes, warmed to room temperature, and stirred overnight. The solution was concentrated and partitioned between ethyl acetate and aqueous Na2CO3 solution. The layers were separated, and the aqueous layer extracted with ethyl acetate. The organic layers were combined, rinsed with aqueous Na2CO3 solution and brine, dried over sodium sulfate, filtered, and concentrated to provide the title compound.
2.32.19 methyl 2-(5-bromo-6-(tert-butoxycarbonyl)pyridin-2-yl)- 1,2,3,4-tetrahydroisoquinoline-8-carboxylate
To a solution of methyl 1,2,3,4-tetrahydroisoquinoline-8-carboxylate hydrochloride (12.37 g) and Example 2.32.18 (15 g) in dimethyl sulfoxide (100 mL) was added N,N-diisopropylethylamine (12 mL). The mixture was stirred at 50oC for 24 hours. The mixture was then diluted with ethyl acetate (500 mL), washed with water and brine, and dried over Na2SO4. Filtration and evaporation of the solvent gave a residue that was purified by silica gel chromatography, eluting with 20% ethyl acetate in heptane, to give the title compound. MS (ESI) m/e 448.4 (M+H)+.
2.32.20 methyl 2-(6-(tert-butoxycarbonyl)-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyridin-2-yl)-1,2,3,4-tetrahydroisoquinoline- 8-carboxylate
To a solution of Example 2.32.19 (2.25 g) and [1,1'- bis(diphenylphosphino)ferrocene]dichloropalladium(II)(205 mg) in acetonitrile (30 mL) was added triethylamine (3 mL) and pinacolborane (2 mL). The mixture was stirred at reflux for 3 hours. The mixture was diluted with ethyl acetate (200 mL) and washed with water and brine, and dried over Na2SO4. Filtration, evaporation of the solvent, and silica gel chromatography (eluted with 20% ethyl acetate in heptane) gave the title compound. MS (ESI) m/e 495.4 (M+H)+.
2.32.21 methyl 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl)-5-methyl-1H- pyrazol-4-yl)pyridin-2-yl)-1,2,3,4-tetrahydroisoquinoline-8- carboxylate
To a solution of Example 2.32.20 (4.94 g) in tetrahydrofuran (60 mL) and water (20 mL) was added Example 2.32.16 (5.57 g), 1,3,5,7-tetramethyl-8-tetradecyl-2,4,6-trioxa-8-phosphaadamantane (412 mg), tris(dibenzylideneacetone)dipalladium(0) (457 mg), and K3PO4 (11 g). The mixture was stirred at reflux for 24 hours. The reaction mixture was cooled, diluted with ethyl acetate (500 mL), washed with water and brine, and dried over Na2SO4. Filtration and evaporation of the solvent gave a residue that was purified by silica gel chromatography, eluting with 20% ethyl acetate in heptane, to give the title compound. MS (ESI) m/e 799.1 (M+H)+. 2.32.22 2-(6-(tert-butoxycarbonyl)-5-(1-((3-(2-((tert- butoxycarbonyl)(methyl)amino)ethoxy)-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl)-5-methyl-1H- pyrazol-4-yl)pyridin-2-yl)-1,2,3,4-tetrahydroisoquinoline-8- carboxylic acid
To a solution of Example 2.32.21 (10 g) in tetrahydrofuran (60 mL), methanol (30 mL) and water (30 mL) was added lithium hydroxide monohydrate (1.2 g). The mixture was stirred at room temperature for 24 hours. The reaction mixture was neutralized with 2% aqueous HCl and concentrated under vacuum. The residue was diluted with ethyl acetate (800 mL) and washed with water and brine, and dried over Na2SO4. Filtration and evaporation of the solvent gave the title compound. MS (ESI) m/e 785.1 (M+H)+.
2.32.23 tert-butyl 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]-3-{1-[(3-{2-[(tert- butoxycarbonyl)(methyl)amino]ethoxy}-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H- pyrazol-4-yl}pyridine-2-carboxylate
To a solution of Example 2.32.22 (10 g) in N,N-dimethylformamide (20 mL) was added benzo[d]thiazol-2-amine (3.24 g), fluoro-N,N,N',N'-tetramethylformamidinium hexafluorophosphate (5.69 g) and N,N-diisopropylethylamine (5.57 g). The mixture was stirred at 60 oC for 3 hours. The reaction mixture was diluted with ethyl acetate (800 mL) and washed with water and brine, and dried over Na2SO4. Filtration and evaporation of the solvent gave a residue that was purified by silica gel chromatography, eluting with 20% ethyl acetate in dichloromethane, to give the title compound. MS (ESI) m/e 915.5 (M+H)+.
2.32.24 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5- methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid
To a solution of Example 2.32.23 (5 g) in dichloromethane (20 mL) was added trifluoroacetic acid (10 mL). The mixture was stirred overnight. The solvent was evaporated under vacuum, and the residue was dissolved in dimethyl sulfoxide/methanol (1:1, 10 mL), and chromatographed via reverse- phase using an Analogix system and a C18 cartridge (300 g), eluting with 10-85% acetonitrile and 0.1% trifluoroacetic acid in water, to give the title compound as a TFA salt. 1H NMR (300 MHz, dimethyl sulfoxide d6) δ ppm 12.85 (s, 1H), 8.13-8.30 (m, 2H), 8.03 (d, 1H), 7.79 (d, 1H), 7.62 (d, 1H), 7.32-7.54 (m, 3H), 7.28 (d, 1H), 6.96 (d, 1H), 4.96 (dd, 1H), 3.80-3.92 (m, 4H), 3.48-3.59 (m, 1H), 2.91-3.11 (m, 2H), 2.51-2.59 (m, 4H), 2.03-2.16 (m, 2H), 1.21-1.49 (m, 6H), 0.97-1.20 (m, 4H), 0.87 (s, 6H). MS (ESI) m/e 760.4 (M+H)+. 2.32.25 3-(1-((3-(2-((((3-(3-aminopropanamido)-4-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)picolinic acid
To a solution of Example 2.32.24 (325 mg) and Example 2.32.7 (382 mg) in N,N- dimethylformamide (9 mL) at 0 oC was added N,N-diisopropylamine (49.1 mg). The reaction mixture was stirred at 0 oC for 5 hours, and acetic acid (22.8 mg) was added. The resulting mixture was diluted with ethyl acetate and washed with water and brine. The organic layer was dried over Na2SO4, filtered and concentrated. The residue was dissolved in a mixture of tetrahydrofuran (10 mL) and methanol (5 mL). To this solution at 0 oC was added 1 M aqueous lithium hydroxide solution (3.8 mL). The resulting mixture was stirred at 0 oC for 1 hour, acidified with acetic acid and concentrated. The concentrate was lyophilized to provide a powder. The powder was dissolved in N,N- dimethylformamide (10 mL), cooled in an ice-bath, and piperidine (1 mL) at 0 oC was added. The mixture was stirred at 0 oC for 15 minutes and 1.5 mL of acetic acid was added. The solution was purified by reverse-phase HPLC using a Gilson system, eluting with 30-80% acetonitrile in water containing 0.1% v/v trifluoroacetic acid, to provide the title compound. MS (ESI) m/e 1172.2 (M+H)+.
2.32.26 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl- 1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-2-({N-[6-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-beta- alanyl}amino)phenyl beta-D-glucopyranosiduronic acid
To Example 2.32.25 (200 mg) in N,N-dimethylformamide (5 mL) at 0 °C was added 2,5- dioxopyrrolidin-1-yl 6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoate (105 mg) and N,N- diisopropylethylamine (0.12 mL). The mixture was stirred at 0 °C for 15 minutes, warmed to room temperature and purified by reverse-phase HPLC on a Gilson system using a 100g C18 column, eluting with 30-80% acetonitrile in water containing 0.1% v/v trifluoroacetic acid, to provide the title compound. 1H NMR (500 MHz, dimethyl sulfoxide-d6) δ ppm 12.85 (s, 2H) 9.07 (s, 1H) 8.18 (s, 1H) 8.03 (d, 1H) 7.87 (t, 1H) 7.79 (d, 1H) 7.61 (d, 1H) 7.41-7.53 (m, 3H) 7.36 (q, 2H) 7.28 (s, 1H) 7.03- 7.09 (m, 1H) 6.96-7.03 (m, 3H) 6.94 (d, 1H) 4.95 (s, 4H) 4.82 (t, 1H) 3.88 (t, 3H) 3.80 (d, 2H) 3.01 (t, 2H) 2.86 (d, 3H) 2.54 (t, 2H) 2.08 (s, 3H) 2.03 (t, 2H) 1.40-1.53 (m, 4H) 1.34 (d, 2H) 0.90-1.28 (m, 12H) 0.82 (d, 6H). MS (ESI) m/e 1365.3 (M+H)+.
2.33 Synthesis of Control Synthon 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-34-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-2-({N-[19-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)-17-oxo-4,7,10,13-tetraoxa-16-azanonadecan-1- oyl]-beta-alanyl}amino)phenyl beta-D-glucopyranosiduronic acid (Synthon I)
The title compound was prepared using the procedure in Example 2.32.26, replacing 2,5- dioxopyrrolidin-1-yl 6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoate with 2,5-dioxopyrrolidin-1- yl 1-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)-3-oxo-7,10,13,16-tetraoxa-4-azanonadecan-19-oate. 1H NMR (500 MHz, dimethyl sulfoxide-d6) δ ppm 8.95 (s, 1H) 8.16 (s, 1H) 7.99 (d, 1H) 7.57-7.81 (m, 4H) 7.38-7.50 (m, 3H) 7.34 (q, 2H) 7.27 (s, 1H) 7.10 (d, 1H) 7.00 (d, 1H) 6.88-6.95 (m, 2H) 4.97 (d, 4H) 4.76 (d, 2H) 3.89 (t, 2H) 3.84 (d, 2H) 3.80 (s, 2H) 3.57-3.63 (m, 4H) 3.44-3.50 (m, 4H) 3.32-3.43 (m, 6H) 3.29 (t, 2H) 3.16 (q, 2H) 3.02 (t, 2H) 2.87 (s, 3H) 2.52-2.60 (m, 2H) 2.29-2.39 (m, 3H) 2.09 (s, 3H) 1.37 (s, 2H) 1.20-1.29 (m, 4H) 1.06-1.18 (m, 4H) 0.92-1.05 (m, 2H) 0.83 (s, 6H). MS (ESI) m/e 1568.6 (M-H)-.
2.34 Synthesis of 4-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)- 1,2,3,4-tetrahydroquinolin-7-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-3-{2-[2-({N-[6-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L- alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid
(Synthon OG)
2.34.1 3-(1-((3-(2-((((2-(2-(2-aminoethoxy)ethoxy)-4- (((2R,3S,4R,5R,6R)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)(2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(1-(benzo[d]thiazol-2- ylcarbamoyl)-1,2,3,4-tetrahydroquinolin-7-yl)picolinic acid
To a cold (0 °C) solution of Example 2.11.6 (279 mg) and Example 1.14.9 (240 mg) in N,N- dimethylformamide (10 mL) was added N,N-diisopropylethylamine (0.157 mL). The reaction was slowly warmed to room temperature and was stirred overnight. To the reaction was added water (2 mL) and LiOH H2O (50 mg), and the mixture was stirred at room temperature for 3 hours. The mixture was acidified with trifluoroacetic acid, filtered and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1%
trifluoroacetic acid, to provide the title compound. MS (ESI) m/e 1233.0 (M-H)-.
2.34.2 3-(1-((3-(2-((((2-(2-(2-((R)-2-amino-3- sulfopropanamido)ethoxy)ethoxy)-4-(((2R,3S,4R,5R,6R)-6- carboxy-345-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(2-methoxyethyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(1-(benzo[d]thiazol-2-ylcarbamoyl)-1,2,3,4- tetrahydroquinolin-7-yl)picolinic acid
To a solution of (R)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3-sulfopropanoic acid (45.7 mg) in N,N-dimethylformamide (1 mL) was added O-(7-azabenzotriazol-1-yl)-N,N,N’,N’- tetramethyluronium hexafluorophosphate (45 mg) and N,N-diisopropylethylamine (0.02 mL). The mixture was stirred at room temperature for 10 minutes, and a solution of Example 2.34.1 (96 mg) and N,N-diisopropylethylamine (0.1 mL) in N,N-dimethylformamide (2 mL) was added. The reaction mixture was stirred at room temperature for 3 hours. To the reaction mixture was added diethylamine (0.1 mL), and the reaction was stirred at room temperature overnight. The mixture was diluted with N,N-dimethylformamide (2 mL), filtered and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1%
trifluoroacetic acid, to provide the title compound. MS (ESI) m/e 1382.2 (M-H)-.
2.34.3 4-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4- tetrahydroquinolin-7-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-3-{2-[2- ({N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3- sulfo-L-alanyl}amino)ethoxy]ethoxy}phenyl beta-D- glucopyranosiduronic acid
The title compound was prepared as described in Example 2.5.3, substituting Example 2.5.2 with Example 2.34.2. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 8.38 (s, 1H), 7.99 (d, 1H), 7.90– 7.70 (m, 6H), 7.44 (s, 1H), 7.35 (t, 1H), 7.28 (d, 1H), 7.24– 7.14 (m, 2H), 6.96 (s, 1H), 6.66 (s, 1H), 5.04 (s, 1H), 4.95 (s, 2H), 4.28 (q, 1H), 4.07 (d, 2H), 3.89 (dd, 3H), 3.22 (ddd, 6H), 2.87– 2.61 (m, 4H), 2.20 (s, 3H), 2.04 (t, 2H), 1.93 (p, 2H), 1.54– 0.90 (m, 20H), 0.83 (d, 7H). MS (ESI) m/e 1575.2 (M-H)-.
2.35 Synthesis of 2-[({[2-({3-[(4-{6-[5-(1,3-benzothiazol-2- ylcarbamoyl)quinolin-3-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-5-{2-[2-({N-[6-(2,5-dioxo- 2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L- alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid
(Synthon OH)
2.35.1 3-(1-((3-(2-((((4-(2-(2-aminoethoxy)ethoxy)-2- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(5-(benzo[d]thiazol-2-ylcarbamoyl)quinolin-3-yl)picolinic acid
To a cold (0 °C) solution of Example 2.26.7 (76 mg) and 6-(5-(benzo[d]thiazol-2- ylcarbamoyl)quinolin-3-yl)-3-(1-((3,5-dimethyl-7-(2-(methylamino)ethoxy)adamantan-1-yl)methyl)- 5-methyl-1H-pyrazol-4-yl)picolinic acid (62 mg) in N,N-dimethylformamide (2 mL) was added N,N- diisopropylethylamine (0.043 mL). The reaction was slowly warmed to room temperature and stirred overnight. To the reaction was added water (2 mL) and LiOH H2O (50 mg), and the mixture was stirred at room temperature for 3 hours. The mixture was acidified with trifluoroacetic acid, filtered and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1% trifluoroacetic acid, to provide the title compound. MS (ESI) m/e 1183.3 (M-H)-.
2.35.2 3-(1-((3-(2-((((4-(2-(2-((R)-2-amino-3- sulfopropanamido)ethoxy)ethoxy)-2-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (5-(benzo[d]thiazol-2-ylcarbamoyl)quinolin-3-yl)picolinic acid
To a solution of (R)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3-sulfopropanoic acid (22.3 mg) in N,N-dimethylformamide (1 mL) was added O-(7-azabenzotriazol-1-yl)-N,N,N’,N’- tetramethyluronium hexafluorophosphate (22 mg) and N,N-diisopropylethylamine (0.02 mL). The mixture was stirred at room temperature for 10 minutes, and a solution of Example 2.35.1 (45 mg) and N,N-diisopropylethylamine (0.1 mL) in N,N-dimethylformamide(2 mL) was added. The reaction was stirred at room temperature for 3 hours. To the reaction mixture was added diethylamine (0.1 mL), and the reaction was stirred at room temperature overnight. The mixture was diluted with N,N-dimethylformamide (2 mL), filtered and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1% trifluoroacetic acid, to provide the title compound. MS (ESI) m/e 1334.5 (M-H)-.
2.35.3 2-[({[2-({3-[(4-{6-[5-(1,3-benzothiazol-2-ylcarbamoyl)quinolin- 3-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1- yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-5-{2-[2-({N-[6- (2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L- alanyl}amino)ethoxy]ethoxy}phenyl beta-D- glucopyranosiduronic acid
The title compound was prepared as described in Example 2.34.1, substituting Example 2.5.2 with Example 2352 1H NMR (400 MHz dimethyl sulfoxide-d6) δ ppm 972 (d 1H) 943 (s 1H) 8.32 (dd, 2H), 8.17 (d, 1H), 8.06 (d, 1H), 8.02– 7.92 (m, 2H), 7.86 (d, 1H), 7.82– 7.71 (m, 2H), 7.52 – 7.43 (m, 2H), 7.36 (t, 1H), 7.17 (d, 1H), 6.96 (s, 2H), 6.69 (d, 1H), 6.58 (dd, 1H), 5.03 (dd, 3H), 4.28 (q, 1H), 4.02 (d, 3H), 3.93 (d, 1H), 3.47– 3.21 (m, 8H), 3.16 (p, 1H), 2.85 (d, 3H), 2.80– 2.63 (m, 2H), 2.22 (s, 3H), 2.04 (t, 2H), 1.53– 1.30 (m, 6H), 1.32– 0.90 (m, 12H), 0.83 (d, 6H). MS (ESI) m/e 1527.4 (M-H)-.
2.36 Synthesis of 2-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)- 1,2,3,4-tetrahydroquinolin-7-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-5-[2-(2-{[6-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)hexanoyl]amino}ethoxy)ethoxy]phenyl beta-D- glucopyranosiduronic acid (Synthon ON)
2.36.1 3-(1-((3-(2-((((4-(2-(2-aminoethoxy)ethoxy)-2- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(1-(benzo[d]thiazol-2-ylcarbamoyl)-1,2,3,4- tetrahydroquinolin-7-yl)picolinic acid, Trifluoroacetic Acid
To a solution of Example 1.1.14 (157 mg) and Example 2.26.7 (167 mg) in N,N- dimethylformamide (3 mL) at 0 °C was added N,N-diisopropylethylamine (188 µL). The mixture was warmed to room temperature, stirred overnight and concentrated. The residue was dissolved in methanol (2 mL) and tetrahydrofuran (3 mL). The solution was cooled in an ice water bath and 1M aqueous lithium hydroxide solution (1.14 mL) was added. The mixture was stirred 0 oC at room temperature for 2 hours and concentrated. The residue was dissolved in dimethyl sulfoxide and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1% trifluoroacetic acid, to provide the title compound.
2.36.2 2-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4- tetrahydroquinolin-7-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-5-[2-(2-{[6-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl]amino}ethoxy)ethoxy]phenyl beta-D- glucopyranosiduronic acid
To a solution of Example 2.36.1 (18 mg) and 2,5-dioxopyrrolidin-1-yl 6-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)hexanoate (6.39 mg) in N,N-dimethylformamide (3 mL) was added N,N- diisopropylethylamine (24 µL). The resulting mixture was stirred for 1 hour and was purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-75% acetonitrile in water containing 01% trifluoroacetic acid to provide the title compound 1H NMR (400 MHz dimethyl sulfoxide-d6) δ 8.36 (s, 1H), 7.97 (d, 1H), 7.85– 7.70 (m, 4H), 7.43 (s, 1H), 7.38– 7.30 (m, 1H), 7.26 (d, 1H), 7.23– 7.10 (m, 2H), 6.95 (s, 2H), 6.65 (d, 1H), 6.56 (dd, 1H), 5.08– 4.94 (m, 3H), 4.02 (dd, 2H), 3.92 (dd, 3H), 3.84 (s, 2H), 3.67 (t, 2H), 3.31– 3.20 (m, 2H), 3.16 (q, 2H), 2.91– 2.74 (m, 6H), 2.18 (s, 3H), 1.99 (t, 2H), 1.91 (p, 2H), 1.51– 1.29 (m, 5H), 1.29– 0.88 (m, 9H), 0.81 (d, 6H). MS (ESI) m/e 1380.2 (M-H)-.
2.37 Synthesis of 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)naphthalen-2-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-{2-[2-({N-[6-(2,5-dioxo- 2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L- alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid
(Synthon OT)
2.37.1 3-(1-((3-(2-((((2-(2-(2-aminoethoxy)ethoxy)-4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)naphthalen-2- yl)picolinic acid
The title compound was prepared by substituting Example 1.6.3 for Example 1.12.10 and Example 2.11.6 for Example 2.26.7 in Example 2.26.8. MS (ESI) m/e 1182.3 (M-H)-.
2.37.2 3-(1-((3-(2-((((2-(2-(2-((R)-2-amino-3- sulfopropanamido)ethoxy)ethoxy)-4-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (8-(benzo[d]thiazol-2-ylcarbamoyl)naphthalen-2-yl)picolinic acid
The title compound was prepared by substituting Example 2.37.1 for Example 2.9.1 in Example 2.18.1. MS (ESI) m/e 1333.3 (M-H)-.
2.37.3 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)naphthalen-2-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-{2-[2-({N-[6- (2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L- alanyl}amino)ethoxy]ethoxy}phenyl beta-D- glucopyranosiduronic acid The title compound was prepared by substituting Example 2.37.2 for Example 2.9.1 in Example 2.9.2. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 9.02 (s, 1H), 8.37 (d, 1H), 8.23 (d, 1H), 8.20 (d, 1H), 8.18 (d, 1H),, 8.06 (d, 1H), 8.01 (d, 1H), 7.94 (d, 1H), 7.87 (br d, 1H), 7.81 (d, 1H), 7.77 (br t, 1H), 7.70 (dd, 1H), 7.48 (dd, 1H), 7.48 (s, 1H), 7.37 (dd, 1H), 7.19 (d, 1H), 6.97 (s, 2H), 6.68 (d, 1H), 6.59 (dd, 1H), 5.06 (br m, 1H), 4.97 (s, 2H), 4.31 (m, 1H), 4.09 (m, 2H), 3.90 (m, 5H), 3.71 (m, 2H), 3.45 (m, 5H), 3.36 (m, 3H), 3.28 (m, 4H), 3.19 (m, 2H), 2.82 (br d, 2H), 2.76 (dd, 2H), 2.23 (s, 3H), 2.06 (t, 2H), 1.52-1.32 (m, 6H), 1.32-0.92 (m, 10H), 0.85 (br s, 6H). MS (ESI) m/e 1526.4 (M-H)-.
2.38 Synthesis of 2-[({[2-({3-[(4-{6-[4-(1,3-benzothiazol-2- ylcarbamoyl)quinolin-6-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-5-[2-(2-{[6-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)hexanoyl]amino}ethoxy)ethoxy]phenyl beta-D- glucopyranosiduronic acid (Synthon OP)
2.38.1 3-(1-((3-(2-((((4-(2-(2-aminoethoxy)ethoxy)-2- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(4-(benzo[d]thiazol-2-ylcarbamoyl)quinolin-6-yl)picolinic acid
The title compound was prepared as described in Example 2.36.1, substituting Example 1.1.14 with Example 1.11.4.
2.38.2 2-[({[2-({3-[(4-{6-[4-(1,3-benzothiazol-2-ylcarbamoyl)quinolin- 6-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1- yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-5-[2-(2-{[6-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl]amino}ethoxy)ethoxy]phenyl beta-D- glucopyranosiduronic acid
The title compound was prepared as described in Example 2.36.2, substituting Example 2.36.1 with Example 2.38.1. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ 9.12 (d, 1H), 8.93 (s, 1H), 8.60 (dd, 1H), 8.27 (d, 1H), 8.21 (d, 1H), 8.07 (d, 1H), 7.97– 7.90 (m, 2H), 7.81 (d, 2H), 7.47 (d, 2H), 7.37 (t, 1H), 7.17 (d, 1H), 6.96 (s, 2H), 6.67 (d, 1H), 6.58 (dd, 1H), 5.11– 4.96 (m, 3H), 4.04 (dd, 2H), 3.92 (d, 1H), 3.86 (s, 2H), 3.40 (q, 5H), 3.34 (t, 2H), 3.31– 3.22 (m, 4H), 3.17 (q, 2H), 2.85 (d, 3H), 2.20 (s, 3H), 2.00 (t, 2H), 1.51– 1.31 (m, 6H), 1.30– 0.88 (m, 13H), 0.82 (d, 6H). MS (ESI) m/e 1400.3 (M+Na)+. 2.39 Synthesis of 4-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)- 1,2,3,4-tetrahydroquinolin-7-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-{2-[2-({N-[6-(2,5-dioxo- 2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L- alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid (Synthon OU)
2.39.1 3-(1-((3-(2-((((2-(2-(2-aminoethoxy)ethoxy)-4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(1-(benzo[d]thiazol-2-ylcarbamoyl)-1,2,3,4- tetrahydroquinolin-7-yl)picolinic acid
The title compound was prepared by substituting Example 1.1.14 for Example 1.12.10 and Example 2.11.6 for Example 2.26.7 in Example 2.26.8. MS (ESI-) m/e 1187.2 (M-H)-.
2.39.2 3-(1-((3-(2-((((2-(2-(2-((R)-2-amino-3- sulfopropanamido)ethoxy)ethoxy)-4-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (1-(benzo[d]thiazol-2-ylcarbamoyl)-1,2,3,4-tetrahydroquinolin- 7-yl)picolinic acid
The title compound was prepared by substituting Example 2.39.1 for Example 2.9.1 in Example 2.18.1. MS (ESI-) m/e 1338.2 (M-H)-.
2.39.3 4-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4- tetrahydroquinolin-7-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-{2-[2-({N-[6- (2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L- alanyl}amino)ethoxy]ethoxy}phenyl beta-D- glucopyranosiduronic acid
The title compound was prepared by substituting Example 2.39.2 for Example 2.9.1 in Example 2.9.2. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 8.39 (br s 1H), 8.00 (d, 1H), 7.86 (d, 2H), 7.81 (d, 1H), 7.77 (d, 2H), 7.48 (v br s, 1H), 7.46 (s, 1H), 7.37 (t, 1H), 7.29 (d, 1H), 7.23 (d, 1H), 7.19 (d, 1H), 6.92 (s, 2H), 6.68 (d, 1H), 6.59 (dd, 1H), 5.06 (br m, 1H), 4.97 (s, 2H), 4.31 (m, 1H), 4.09 (m, 2H), 3.96 (br t, 2H), 3.88 (br m, 2H), 3.71 (m, 2H), 3.45 (m, 5H), 3.37 (m, 3H), 3.28 (m, 4H), 3.18 (m, 2H), 2.86 (br m, 5H), 2.75 (dd, 2H), 2.22 (s, 3H), 2.06 (t, 2H), 1.95 (m, 2H), 1.52- 1.32 (m, 6H), 1.32-0.92 (m, 12H), 0.85 (br s, 6H). MS (ESI-) m/e 1531.2 (M-H)-.
2.40 Synthesis of 4-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)- 1,2,3,4-tetrahydroquinolin-7-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-(3-{[6-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)hexanoyl]amino}propoxy)phenyl beta-D- glucopyranosiduronic acid (Synthon OO)
2.40.1 3-(1-((3-(2-((((2-(3-aminopropoxy)-4-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (1-(benzo[d]thiazol-2-ylcarbamoyl)-1,2,3,4-tetrahydroquinolin- 7-yl)picolinic acid
The title compound was prepared as described in Example 2.36.1, substituting Example 2.26.7 with Example 2.28.3. MS (ESI) m/e 1159.2 (M+H)+.
2.40.2 4-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4- tetrahydroquinolin-7-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-(3-{[6-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl]amino}propoxy)phenyl beta-D- glucopyranosiduronic acid
The title compound was prepared as described in Example 2.36.2, substituting Example 2.36.1 with Example 2.40.1. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ 8.38 (s, 1H), 7.98 (d, 1H), 7.87– 7.72 (m, 2H), 7.44 (s, 1H), 7.35 (t, 1H), 7.28 (d, 1H), 7.19 (dd, 2H), 6.96 (s, 2H), 6.62 (d, 1H), 6.57 (dd, 1H), 5.03 (s, 1H), 4.95 (s, 2H), 4.03– 3.81 (m, 8H), 3.42– 3.20 (m, 7H), 3.16 (q, 2H), 2.90 – 2.75 (m, 5H), 2.20 (s, 3H), 2.01 (t, 2H), 1.97– 1.87 (m, 2H), 1.80 (t, 2H), 1.45 (td, 4H), 1.13 (d, 8H), 0.83 (d, 6H). MS (ESI) m/e 1350.2 (M-H)-.
2.41 Synthesis of 4-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)- 1,2,3,4-tetrahydroquinolin-7-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-[3-({N-[6-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L- alanyl}amino)propoxy]phenyl beta-D-glucopyranosiduronic acid (Synthon OQ) 2.41.1 3-(1-((3-(2-((((2-(3-((R)-2-amino-3- sulfopropanamido)propoxy)-4-(((2S,3R,4S,5S,6S)-6-carboxy- 3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (1-(benzo[d]thiazol-2-ylcarbamoyl)-1,2,3,4-tetrahydroquinolin- 7-yl)picolinic acid
To a solution of (R)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3-sulfopropanoic acid (35.4 mg) and O-(7-azabenzotriazol-1-yl)-N,N,N’,N’-tetramethyluronium hexafluorophosphate (29.8 mg) in N,N-dimethylformamide (1 mL) at 0 oC was added N,N-diisopropylethylamine (30 µL). The resulting mixture was stirred for 15 minutes and added to a mixture of Example 2.40.1 (70 mg) and N,N-diisopropylethylamine (80 µL) in N,N-dimethylformamide (2 mL). The resulting mixture was stirred for 1 hour. Diethylamine (62.2 µL) was added, and the mixture was stirred for 1 hour. The reaction was cooled in ice-bath and trifluoroacetic acid (93 µL) was added. The mixture was diluted with dimethyl sulfoxide (5.5 mL) and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-75% acetonitrile in water containing 0.1% trifluoroacetic acid, to provide the title compound.
2.41.2 4-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4- tetrahydroquinolin-7-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-[3-({N-[6-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L- alanyl}amino)propoxy]phenyl beta-D-glucopyranosiduronic acid
The title compound was prepared as described in Example 2.36.2, substituting Example 2.36.1 with Example 2.41.1. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ 8.37 (s, 1H), 7.98 (d, 1H), 7.87– 7.72 (m, 5H), 7.44 (s, 1H), 7.35 (t, 1H), 7.27 (d, 1H), 7.20 (t, 1H), 7.16 (d, 1H), 6.96 (s, 2H), 6.63 (d, 1H), 6.55 (dd, 1H), 5.02 (s, 1H), 4.95 (s, 2H), 4.26 (q, 1H), 4.04– 3.79 (m, 8H), 3.32– 3.08 (m, 4H), 2.89– 2.66 (m, 7H), 2.35 (q, 0H), 2.20 (s, 3H), 2.03 (t, 2H), 1.93 (p, 2H), 1.80 (t, 2H), 1.52– 1.30 (m, 6H), 1.30– 0.89 (m, 13H), 0.83 (d, 6H). MS (ESI) m/e 1502.2 (M-H)-.
2.42 Synthesis of 2-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)- 1,2,3,4-tetrahydroquinolin-7-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-5-{2-[2-({N-[6-(2,5-dioxo- 2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L- alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid
(Synthon OR) 2.42.1 3-(1-((3-(2-((((4-(2-(2-((R)-2-amino-3- sulfopropanamido)ethoxy)ethoxy)-2-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (1-(benzo[d]thiazol-2-ylcarbamoyl)-1,2,3,4-tetrahydroquinolin- 7-yl)picolinic acid
The title compound was prepared as described in Example 2.41.1, substituting Example 2.40.1 with Example 2.36.1. MS (ESI) m/e 1338.2 (M-H)-.
2.42.2 2-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4- tetrahydroquinolin-7-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-5-{2-[2-({N-[6- (2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L- alanyl}amino)ethoxy]ethoxy}phenyl beta-D- glucopyranosiduronic acid
The title compound was prepared as described in Example 2.36.2, substituting Example 2.36.1 with Example 2.42.1. 1H NMR (500 MHz, dimethyl sulfoxide-d6) δ 8.39 (s, 1H), 8.00 (d, 1H), 7.86 (t, 2H), 7.83– 7.73 (m, 3H), 7.45 (s, 1H), 7.40– 7.32 (m, 1H), 7.29 (d, 1H), 7.26– 7.13 (m, 2H), 6.97 (s, 2H), 6.70 (d, 1H), 6.59 (dd, 1H), 5.11– 4.94 (m, 3H), 4.29 (dt, 1H), 4.04 (dd, 2H), 3.99– 3.91 (m, 3H), 3.87 (d, 2H), 3.69 (t, 2H), 3.40– 3.07 (m, 7H), 2.91– 2.74 (m, 6H), 2.69 (dd, 1H), 2.21 (s, 3H), 2.05 (t, 2H), 1.94 (p, 2H), 1.53– 1.32 (m, 5H), 1.31– 0.90 (m, 7H), 0.84 (d, 6H). MS (ESI) m/e 1531.2 (M-H)-.
2.43 Synthesis of 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)naphthalen-2-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-3-{2-[2-({N-[6-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L- alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid
(Synthon OS)
2.43.1 3-(1-((3-(2-((((2-(2-(2-aminoethoxy)ethoxy)-4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)(2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)naphthalen-2-yl)picolinic acid The title compound was prepared as described in Example 2.34.1, substituting Example 2.5.2 with Example 1.15.1. MS (ESI) m/e 1228.1 (M-H)-.
2.43.2 3-(1-((3-(2-((((2-(2-(2-((R)-2-amino-3- sulfopropanamido)ethoxy)ethoxy)-4-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(2-methoxyethyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)naphthalen-2- yl)picolinic acid
The title compound was prepared as described in Example 2.34.2, substituting Example 2.34.1 with Example 2.43.2. MS (ESI) m/e 1379.1.1 (M+H)+.
2.43.3 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)naphthalen-2-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2- methoxyethyl)carbamoyl}oxy)methyl]-3-{2-[2-({N-[6-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L- alanyl}amino)ethoxy]ethoxy}phenyl beta-D- glucopyranosiduronic acid
The title compound was prepared as described in Example 2.34, substituting Example 2.34.2 with Example 2.43.2. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 9.00 (s, 1H), 8.36 (d, 1H), 8.27– 8.12 (m, 3H), 8.05 (d, 1H), 8.00 (d, 1H), 7.92 (d, 1H), 7.85 (d, 1H), 7.79 (d, 1H), 7.75 (t, 1H), 7.69 (t, 1H), 7.52– 7.43 (m, 2H), 7.35 (t, 1H), 7.24– 7.12 (m, 1H), 6.95 (s, 2H), 6.66 (s, 1H), 6.57 (d, 1H), 5.04 (d, 1H), 4.95 (s, 2H), 4.29 (q, 1H), 4.15– 4.01 (m, 2H), 3.86 (d, 3H), 3.46– 3.11 (m, 16H), 2.84– 2.62 (m, 2H), 2.21 (d, 3H), 2.04 (t, 2H), 1.53– 1.30 (m, 6H), 1.28– 0.89 (m, 6H), 0.82 (d, 7H). MS (ESI) m/e 1570.4 (M-H)-.
2.44 Synthesis of N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-L- valyl-N-[4-({[{2-[{8-(1,3-benzothiazol-2-ylcarbamoyl)-2-[6-carboxy-5-(1- {[3-(2-methoxyethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}- 5-methyl-1H-pyrazol-4-yl)pyridin-2-yl]-1,2,3,4-tetrahydroisoquinolin-6- yl}(methyl)amino]ethyl}(methyl)carbamoyl]oxy}methyl)phenyl]-L- alaninamide (Synthon OX)
The title compound was prepared as described in Example 2.30, substituting Example 1.17.10 with Example 1.21.12. MS (ESI) m/e 1359.5 (M+H)+, 1357.5 (M-H)-.
2.45 Synthesis of N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-L- valyl-N-{4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-6- methoxy-34-dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]phenyl}-L-alaninamide
(Synthon OZ)
The title compound was prepared as described in Example 2.30, substituting Example 1.17.10 with Example 1.22.9. MS (ESI) m/e 1302.5 (M+H)+, 1300.5 (M-H)-.
2.46 Synthesis of 2-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)naphthalen-2-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-methoxyethyl)carbamoyl}oxy)methyl]-5-{2-[2-({N-[6-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L- alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid
(Synthon PA)
2.46.1 3-(1-((3-(2-((((4-(2-(2-aminoethoxy)ethoxy)-2- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)(2- methoxyethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)naphthalen-2-yl)picolinic acid
The title compound was prepared as described in Example 2.43.1, substituting Example 2.11.6 with Example 2.26.7. MS (ESI) m/e 1228.1 (M-H)-.
2.46.2 3-(1-((3-(2-((((4-(2-(2-((R)-2-amino-3- sulfopropanamido)ethoxy)ethoxy)-2-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(2-methoxyethyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)naphthalen-2- yl)picolinic acid
The title compound was prepared as described in Example 2.34.2, substituting Example 2.34.1 with Example 2.46.1. MS (ESI) m/e 1377.5 (M-H)-.
2.46.3 2-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)naphthalen-2-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2- methoxyethyl)carbamoyl}oxy)methyl]-5-{2-[2-({N-[6-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L- alanyl}amino)ethoxy]ethoxy}phenyl beta-D- glucopyranosiduronic acid The title compound was prepared as described in Example 2.34, substituting Example 2.34.2 with Example 2.46.2. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ ppm 13.08 (s, 1H), 9.00 (s, 1H), 8.36 (d, 1H), 8.25– 8.12 (m, 3H), 8.05 (d, 1H), 8.00 (d, 1H), 7.92 (d, 1H), 7.85 (d, 1H), 7.78 (dd, 2H), 7.72– 7.65 (m, 1H), 7.50– 7.43 (m, 2H), 7.35 (t, 1H), 7.21– 7.14 (m, 1H), 6.96 (s, 2H), 6.69 (d, 1H), 6.58 (d, 1H), 5.13– 4.93 (m, 3H), 4.28 (q, 1H), 4.03 (dd, 2H), 3.94 (d, 1H), 3.86 (d, 2H), 3.67 (t, 2H), 3.31– 3.08 (m, 8H), 2.83– 2.64 (m, 2H), 2.21 (d, 3H), 2.04 (t, 2H), 1.53– 1.30 (m, 5H), 1.30– 0.89 (m, 11H), 0.89– 0.75 (m, 6H). MS (ESI) m/e 1570.5 (M-H)-.
2.47 Synthesis of 2-[({[2-({3-[(4-{6-[5-(1,3-benzothiazol-2- ylcarbamoyl)quinolin-3-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-5-[2-(2-{[6-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)hexanoyl]amino}ethoxy)ethoxy]phenyl beta-D- glucopyranosiduronic acid (Synthon QL)
2.47.1 3-(1-((3-(2-((((4-(2-(2-aminoethoxy)ethoxy)-2- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(5-(benzo[d]thiazol-2-ylcarbamoyl)quinolin-3-yl)picolinic acid
The title compound was prepared as described in Example 2.36.1, substituting Example 1.1.14 with Example 1.10.3.
2.47.2 2-[({[2-({3-[(4-{6-[5-(1,3-benzothiazol-2-ylcarbamoyl)quinolin- 3-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1- yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-5-[2-(2-{[6-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl]amino}ethoxy)ethoxy]phenyl beta-D- glucopyranosiduronic acid
The title compound was prepared as described in Example 2.36., substituting Example 2.36.1 with Example 2.47.1. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ 13.17 (s, 1H), 9.70 (d, 1H), 9.39 (s, 1H), 8.31 (dd, 2H), 8.16 (d, 1H), 8.06 (dd, 1H), 8.01– 7.90 (m, 2H), 7.83– 7.71 (m, 2H), 7.52– 7.43 (m, 2H), 7.39– 7.31 (m, 1H), 7.18 (d, 1H), 6.96 (s, 2H), 6.65 (d, 1H), 6.58 (dd, 1H), 5.04 (s, 1H), 4.96 (s, 2H), 4.09 (dtd, 2H), 3.87 (s, 2H), 3.70 (t, 2H), 3.40– 3.14 (m, 7H), 2.85 (d, 3H), 2.22 (s, 3H), 2.01 (t, 2H), 1.49– 1.30 (m, 6H), 1.30– 0.90 (m, 10H), 0.90– 0.74 (m, 6H). MS (ESI) m/e 1400.4 (M+Na)+.
2.48 Synthesis of 4-[({[2-({3-[(4-{6-[5-(1,3-benzothiazol-2- ylcarbamoyl)quinolin-3-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-[2-(2-{[6-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)hexanoyl]amino}ethoxy)ethoxy]phenyl beta-D- glucopyranosiduronic acid (Synthon QM)
2.48.1 3-(1-((3-(2-((((2-(2-(2-aminoethoxy)ethoxy)-4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(5-(benzo[d]thiazol-2-ylcarbamoyl)quinolin-3-yl)picolinic acid
To a solution of Example 1.10.3 (208 mg) and Example 2.11.6 (267 mg) in N,N- dimethylformamide (2 mL) at 0 °C was added N,N-diisopropylethylamine (251 µL). The resulting mixture was stirred at room temperature overnight and concentrated. The residue was dissolved in methanol (3 mL) and tetrahydrofuran (5 mL). The solution was cooled in an ice water bath and 1M aqueous lithium hydroxide solution was added (2.87 mL). The mixture was stirred at 0 oC for 2 hours and was acidified with trifluoroacetic acid. The reaction mixture was concentrated under reduced pressure. The residue was diluted with dimethyl sulfoxide and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-75% acetonitrile in water containing 0.1%
trifluoroacetic acid, to provide the title compound. MS (ESI) m/e 1185.1 (M+H)+.
2.48.2 4-[({[2-({3-[(4-{6-[5-(1,3-benzothiazol-2-ylcarbamoyl)quinolin- 3-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1- yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-[2-(2-{[6-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl]amino}ethoxy)ethoxy]phenyl beta-D- glucopyranosiduronic acid
The title compound was prepared as described in Example 2.36.2, substituting Example 2.36.1 with Example 2.48.1. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ 13.18 (s, 1H), 9.70 (d, 1H), 9.39 (s, 1H), 8.31 (dd, 2H), 8.16 (d, 1H), 8.06 (d, 1H), 8.01– 7.90 (m, 2H), 7.80 (d, 2H), 7.52– 7.43 (m, 2H), 7.39– 7.32 (m, 1H), 7.18 (d, 1H), 6.96 (s, 2H), 6.67 (d, 1H), 6.58 (dd, 1H), 5.11– 4.90 (m, 3H), 4.03 (d, 2H), 3.95– 3.82 (m, 3H), 3.68 (t, 2H), 3.48– 3.23 (m, 10H), 3.18 (t, 2H), 2.85 (d, 3H), 2.22 (s, 3H), 2.00 (t, 2H), 1.51– 1.31 (m, 5H), 1.19 (dd, 10H), 0.83 (d, 6H). MS (ESI) m/e 1376.4 (M-H)-.
2.49 Synthesis of 6-[5-(1,3-benzothiazol-2-ylcarbamoyl)quinolin-3-yl]-3-(1-{[3- (2-{[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl](methyl)amino}ethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec- 1-yl]methyl}-5-methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid (Synthon QN)
The title compound was prepared as described in Example 2.36.2, substituting Example 2.36.1 with Example 1.10.3. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ 13.21 (s, 1H), 9.70 (d, 1H), 9.40 (s, 1H), 8.42– 8.27 (m, 2H), 8.16 (d, 1H), 8.06 (d, 1H), 8.04– 7.90 (m, 2H), 7.80 (d, 1H), 7.56– 7.44 (m, 2H), 7.42– 7.31 (m, 1H), 6.95 (d, 2H), 3.87 (s, 2H), 3.55– 3.18 (m, 5H), 2.95 (s, 1H), 2.76 (s, 2H), 2.28 (t, 1H), 2.22 (s, 4H), 1.53– 1.29 (m, 6H), 1.28– 0.91 (m, 10H), 0.84 (s, 6H). MS (ESI) m/e 949.1 (M+H)+.
2.50 Synthesis of 4-[({[2-({3-[(4-{6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-1H- indol-2-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]- 5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-2-({N-[3-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)propanoyl]-beta-alanyl}amino)phenyl beta-D- glucopyranosiduronic acid (Synthon QT)
2.50.1 3-(1-((3-(2-((((3-(3-aminopropanamido)-4-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (7-(benzo[d]thiazol-2-ylcarbamoyl)-1H-indol-2-yl)picolinic acid
The title compound was prepared by substituting Example 1.27.4 for Example 2.32.24 in Example 2.32.25. MS (ESI) m/e:1156.6 (M+H)+.
2.50.2 4-[({[2-({3-[(4-{6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-1H- indol-2-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1- yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-2-({N-[3-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)propanoyl]-beta- alanyl}amino)phenyl beta-D-glucopyranosiduronic acid
The title compound was prepared by substituting Example 2.50.1 for Example 2.11.7 in Example 2.11.8. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ ppm 13.00 (s, 2H); 9.06 (s, 1H), 8.29 (dd, 1H), 8.22 (d, 1H), 8.18 (s, 1H), 8.04 (t, 2H), 7.97 (d, 1H), 7.90 (d, 1H), 7.79 (d, 1H), 7.50– 7.43 (m, 3H), 7.35 (ddd, 1H), 7.25 (t, 1H), 7.06 (d, 1H), 7.01 (dd, 1H), 6.94 (s, 2H), 4.96 (s, 2H), 4.81 (s, 1H), 3.33– 3.25 (m, 6H), 2.87 (d, 3H), 2.50 (d, 3H), 2.31 (dd, 2H), 2.21 (s, 3H), 1.38 (d, 2H), 1.30– 0.77 (m, 18H). MS (ESI) m/e 1305.2 (M-H)-.
2.51 Synthesis of 4-[({[2-({3-[(4-{6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-1H- indol-2-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]- 5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-[2-(2-{[3-(25-dioxo-25- dihydro-1H-pyrrol-1-yl)propanoyl]amino}ethoxy)ethoxy]phenyl beta-D- glucopyranosiduronic acid (Synthon RF)
2.51.1 3-(1-((3-(2-((((2-(2-(2-aminoethoxy)ethoxy)-4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(7-(benzo[d]thiazol-2-ylcarbamoyl)-1H-indol-2- yl)picolinic acid
The title compound was prepared by substituting Example 1.27.4 for Example 1.12.10 in Example 2.11.7. MS (ESI) m/e:1172.9 (M+H)+. 2.51.2 4-[({[2-({3-[(4-{6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-1H- indol-2-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1- yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-[2-(2-{[3-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1- yl)propanoyl]amino}ethoxy)ethoxy]phenyl beta-D- glucopyranosiduronic acid
The title compound was prepared by substituting Example 2.51.1 for Example 2.11.7 in Example 2.11.8. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ ppm 11.16 (s, 2H), 8.27 (d, 1H), 8.19 (d, 1H), 8.06– 7.94 (m, 3H), 7.88 (d, 1H), 7.77 (d, 1H), 7.50– 7.39 (m, 3H), 7.33 (t, 1H), 7.26– 7.13 (m, 2H), 6.93 (s, 2H), 6.63 (d, 1H), 6.57 (dd, 1H), 5.03 (d, 1H), 4.94 (s, 2H), 4.13– 4.00 (m, 2H), 3.86 (d, 3H), 3.14 (q, 2H), 2.83 (d, 3H), 2.29 (t, 2H), 2.20 (s, 3H), 1.36 (d, 2H), 1.28– 0.73 (m, 16H). MS (ESI) m/e 1322.4 (M-H)-.
2.52 Synthesis of 4-[({[2-({3-[(4-{6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-1H- indol-2-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]- 5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-{2-[2-({N-[3-(2,5-dioxo- 2,5-dihydro-1H-pyrrol-1-yl)propanoyl]-3-sulfo-L- alanyl}amino)ethoxy]ethoxy}phenyl beta-D-glucopyranosiduronic acid
(Synthon RG)
2.52.1 3-(1-((3-(2-((((2-(2-(2-((R)-2-amino-3- sulfopropanamido)ethoxy)ethoxy)-4-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (7-(benzo[d]thiazol-2-ylcarbamoyl)-1H-indol-2-yl)picolinic acid The title compound was prepared by substituting Example 2.51.1 for Example 2.9.1 in Example 2.18.1. MS (ESI) m/e:1325.5 (M+H)+.
2.52.2 4-[({[2-({3-[(4-{6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-1H- indol-2-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1- yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-{2-[2-({N-[3- (2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)propanoyl]-3-sulfo-L- alanyl}amino)ethoxy]ethoxy}phenyl beta-D- glucopyranosiduronic acid
The title compound was prepared by substituting Example 2.52.1 for Example 2.11.7 in Example 2.11.8. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ ppm 11.17 (s, 2H), 8.27 (d, 1H), 8.20 (d, 1H), 8.03 (dd, 2H), 7.96 (d, 1H), 7.89 (d, 1H), 7.82– 7.75 (m, 2H), 7.50 (s, 1H), 7.48– 7.41 (m, 2H), 7.34 (t, 1H), 7.24 (t, 1H), 7.18 (d, 1H), 6.93 (s, 2H), 6.66 (d, 1H), 6.58 (dd, 1H), 5.04 (d, 1H), 4.95 (s, 2H), 3.70 (t, 2H), 3.58 (t, 2H), 3.48– 3.14 (m, 11H), 2.89– 2.79 (m, 4H), 2.73 (dd, 1H), 2.37 (m, 2H), 2.21 (s, 3H), 1.45– 0.73 (m, 19H). MS (ESI) m/e 1473.3 (M-H)-.
2.53 Synthesis of 4-[({[2-({3-[(4-{6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-3- methyl-1H-indol-2-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1- yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-[2-(2-{[3-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)propanoyl]amino}ethoxy)ethoxy]phenyl beta-D- glucopyranosiduronic acid (Synthon SF)
2.53.1 3-(1-((3-(2-((((2-(2-(2-aminoethoxy)ethoxy)-4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(7-(benzo[d]thiazol-2-ylcarbamoyl)-3-methyl-1H-indol-2- yl)picolinic acid
The title compound was prepared by substituting Example 1.29.7 for Example 1.12.10 in Example 2.11.7. MS (ESI) m/e:1187.1 (M+H)+. 2.53.2 4-[({[2-({3-[(4-{6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-3- methyl-1H-indol-2-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-[2-(2-{[3-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1- yl)propanoyl]amino}ethoxy)ethoxy]phenyl beta-D- glucopyranosiduronic acid The title compound was prepared by substituting Example 2.53.1 for Example 2.11.7 in Example 2.11.8. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ ppm 11.01 (s, 1H), 8.28 (d, 1H), 8.06 – 7.94 (m, 4H), 7.91 (d, 1H), 7.76 (d, 1H), 7.50– 7.42 (m, 2H), 7.32 (td, 1H), 7.26– 7.15 (m, 2H), 6.93 (s, 2H), 6.64 (d, 1H), 6.58 (dd, 1H), 5.03 (d, 1H), 4.95 (s, 2H), 4.11– 3.99 (m, 2H), 3.87 (d, 3H), 3.68 (t, 2H), 3.56 (dd, 2H), 3.47– 3.33 (m, 5H), 3.33– 3.19 (m, 4H), 3.14 (q, 2H), 2.84 (d, 3H), 2.63 (s, 3H), 2.30 (dd, 2H), 2.21 (s, 3H), 1.42– 0.72 (m, 21H). MS (ESI) m/e 1336.3 (M-H)-.
2.54 Synthesis of N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-L- valyl-N-{4-[({[2-({3-[(4-{6-[4-(1,3-benzothiazol-2- ylcarbamoyl)isoquinolin-6-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]phenyl}-N5-carbamoyl-L- ornithinamide (Synthon SR)
The title compound was prepared as described in Example 2.2, substituting Example 1.3.2 with Example 1.26.10. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 13.28 (s, 2H), 9.96 (s, 1H), 9.59 (s, 1H), 9.03 (d, 2H), 8.53 (d, 1H), 8.42 (d, 1H), 8.25 (d, 1H), 8.05 (t, 2H), 7.97 (d, 1H), 7.78 (dd, 2H), 7.58 (d, 2H), 7.47 (d, 2H), 7.36 (t, 1H), 7.26 (d, 2H), 6.97 (s, 2H), 5.96 (s, 1H), 4.96 (s, 2H), 4.45– 4.29 (m, 1H), 4.17 (t, 1H), 3.51– 3.18 (m, 6H), 3.07– 2.75 (m, 4H), 2.22 (s, 3H), 2.11 (dq, 1H), 2.02– 1.82 (m, 1H), 1.76– 0.88 (m, 18H), 0.81 (dd, 14H). MS (ESI) m/e 1352.4 (M-H)-.
2.55 Synthesis of 4-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-5,6- dihydroimidazo[1,5-a]pyrazin-7(8H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl]carbamoyl}oxy)methyl]-3-[2-(2-{[(2,5-dioxo-2,5-dihydro- 1H-pyrrol-1-yl)acetyl]amino}ethoxy)ethoxy]phenyl beta-D- glucopyranosiduronic acid (Synthon YZ)
2.55.1 3-(1-((3-(2-((((2-(2-(2-aminoethoxy)ethoxy)-4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (1-(benzo[d]thiazol-2-ylcarbamoyl)-5,6-dihydroimidazo[1,5- a]pyrazin-7(8H)-yl)picolinic acid
The title compound was prepared by substituting Example 1.4.10 for Example 1.12.10 in Example 2.11.7. MS (ESI) m/e 1165 (M+H)+, 1163 (M-H)-.
2.55.2 4-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-5,6- dihydroimidazo[1,5-a]pyrazin-7(8H)-yl]-2-carboxypyridin-3- yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl]carbamoyl}oxy)methyl]-3-[2-(2-{[(25-dioxo-25- dihydro-1H-pyrrol-1-yl)acetyl]amino}ethoxy)ethoxy]phenyl
beta-D-glucopyranosiduronic acid
The title compound was prepared by substituting Example 2.55.1 for Example 2.9.1 in Example 2.10. 1H NMR (300 MHz, dimethyl sulfoxide-d6) δ ppm 8.22 (t, 1H), 8.05 (s, 1H), 7.99 (d, 1H), 7.76 (d, 1H), 7.61 (d, 1H), 7.46 (t, 1H), 7.35-7.31 (m, 2H), 7.20 (d, 1H), 7.15 (d, 1H), 7.07 (s, 2H), 6.66 (d, 1H), 6.61 (dd, 1H), 5.12 (s, 2H), 5.08 (d, 1H), 4.94 (s, 2H), 4.28 (t, 2H), 4.09 (m, 4H), 4.03 (s, 2H), 3.91 (m, 3H), 3.84 (m, 4H), 3.73 (t, 2H), 3.49 (t, 2H), 3.40 (t, 2H), 3.34 (m, 2H), 3.30 (dd, 2H), 3.26 (m, 2H), 3.06 (q, 2H), 2.13 (s, 3H), 1.39 (bs, 2H), 1.26 (q, 4H), 1.13 (q, 4H), 1.02 (q, 2H), 0.85 (s, 6H). MS (ESI) m/e 1302 (M+H)+.
2.56 Synthesis of 2-[({[2-({3-[(4-{6-[5-(1,3-benzothiazol-2- ylcarbamoyl)quinolin-3-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl]carbamoyl}oxy)methyl]-4-[19-(2,5-dioxo-2,5-dihydro-1H- pyrrol-1-yl)-14-oxo-4,7,10-trioxa-13-azanonadec-1-yl]phenyl beta-D- glucopyranosiduronic acid (Synthon QR)
2.56.1 3-(1-((3-(2-((((5-(3-(2-(2-(2- aminoethoxy)ethoxy)ethoxy)propyl)-2-(((3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (5-(benzo[d]thiazol-2-ylcarbamoyl)quinolin-3-yl)picolinic acid
To a cold (0 °C) solution of (3R,4S,5S,6S)-2-(4-(1-(9H-fluoren-9-yl)-3-oxo-2,7,10,13- tetraoxa-4-azahexadecan-16-yl)-2-((((4-nitrophenoxy)carbonyl)oxy)methyl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (56 mg) and Example 1.43.5 (47 mg) in N,N-dimethylformamide (2 mL) was added N,N-diisopropylethylamine (0.026 mL). The reaction was slowly warmed to room temperature and stirred overnight. To the reaction was added water (2 mL) and LiOH H2O (50 mg), and the mixture was stirred at room temperature for 3 hours. The mixture was acidified with trifluoroacetic acid, filtered and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1%
trifluoroacetic acid, to provide the title compound.
MS (ESI) m/e 1255.4 (M-H)-.
2.56.2 2-[({[2-({3-[(4-{6-[5-(1,3-benzothiazol-2-ylcarbamoyl)quinolin- 3-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1- yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl]carbamoyl}oxy)methyl]-4-[19-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)-14-oxo-4,7,10-trioxa-13-azanonadec- 1-yl]phenyl beta-D-glucopyranosiduronic acid To a solution of Example 2.56.1 (21 mg) in N,N-dimethylformamide (2 mL) was added 2,5- dioxopyrrolidin-1-yl 6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoate (5.24 mg) and N,N- diisopropylethylamine (0.012 mL). The reaction mixture was stirred at room temperature overnight. The mixture was diluted with N,N-dimethylformamide (2 mL), filtered and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1% trifluoroacetic acid, to provide the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 13.17 (s, 2H), 9.68 (d, 1H), 9.37 (s, 1H), 8.29 (dd, 2H), 8.14 (d, 1H), 8.04 (d, 1H), 8.01– 7.88 (m, 2H), 7.82– 7.69 (m, 2H), 7.51– 7.40 (m, 2H), 7.38– 7.29 (m, 1H), 7.17 (t, 1H), 7.13– 7.01 (m, 2H), 6.95 (s, 3H), 5.02 (s, 2H), 4.94– 4.86 (m, 1H), 3.91– 3.79 (m, 4H), 3.33 (td, 9H), 3.29– 3.22 (m, 2H), 3.12 (q, 2H), 3.04 (d, 2H), 2.20 (s, 3H), 1.98 (t, 2H), 1.70 (p, 2H), 1.42 (dt, 7H), 1.31– 0.89 (m, 13H), 0.82 (s, 7H). MS (ESI) m/e 1448.3 (M-H)-.
2.57 Synthesis of 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)naphthalen-2-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-[4-({N-[6-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L-alanyl}amino)butyl]phenyl beta-D-glucopyranosiduronic acid (Synthon SE)
2.57.1 (2S,3R,4S,5S,6S)-2-(3-bromo-4-formylphenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
A mixture of (3R,4S,5S,6S)-2-bromo-6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate (2.67 g), 2-bromo-4-hydroxybenzaldehyde (0.90 g) and silver oxide (1.56 g) was stirred in acetonitrile (20 mL) at room temperature protected from light. After 3 hours, the reaction was diluted with dichloromethane (20 mL), filtered through diatomaceous earth, washed with additional dichloromethane (40 mL) and concentrated. The residue was purified by silica gel chromatography, eluting with a gradient of 5% to 50% hexanes/ethyl acetate over 30 minutes, to provide the title compound. MS (ESI) m/e 517.1 (M+H)+.
2.57.2 (9H-fluoren-9-yl)methyl but-3-yn-1-ylcarbamate
A solution of but-3-yn-1-amine hydrochloride (9 g) and N-ethyl-N-isopropylpropan-2-amine (44.7 mL) was stirred in dichloromethane(70 mL) and the mixture was cooled to 0 °C. A solution of (9H-fluoren-9-yl)methyl carbonochloridate (22.06 g) in dichloromethane (35 mL) was added, and the reaction was stirred for 2 hours. The reaction mixture was concentrated. The crude material was deposited onto silica gel, loaded onto a silica gel column and eluted with petroleum diethyl ether/ethyl acetate (10%-25%) to provide the title compound. MS (ESI) m/e 314 (M+Na)+.
2.57.3 (2S,3R,4S,5S,6S)-2-(3-(4-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)but-1-yn-1-yl)-4-formylphenoxy)- 6-(methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate Example 2.57.1 (0.389 g), Example 2.57.2 (0.285 g), bis(triphenylphosphsine)palladium(II) dichloride (0.053 g), and copper(I) iodide (0.014 g) were weighed into a vial and the vial was flushed with a stream of nitrogen. N,N-diisopropylethylamine (0.263 mL) and N,N-dimethylformamide (1.5 mL) were added, and the reaction was stirred at room temperature overnight. The reaction mixture was diluted with diethyl ether (50 mL) and washed with water (30 mL) and brine (30 mL). The organic layer was dried over magnesium sulfate, filtered, and concentrated. The residue was purified by silica gel chromatography, eluting with a gradient of 5% to 60% ethyl acetate/heptanes over 30 minutes, to provide the title compound. MS (ESI) m/e 728.4 (M+H)+.
2.57.4 (2S,3R,4S,5S,6S)-2-(3-(4-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)butyl)-4-formylphenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
Example 2.57.3 (262 mg) and tetrahydrofuran (10 mL) were added to10% palladium/C (50 mg) in a 50 mL pressure bottle and the mixture was shaken for 2 hours at room temperature under 30 psi H2. The reaction mixture was filtered and concentrated to provide the title compound. MS (ESI) m/e 732.5 (M+H)+.
2.57.5 (2S,3R,4S,5S,6S)-2-(3-(4-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)butyl)-4- (hydroxymethyl)phenoxy)-6-(methoxycarbonyl)tetrahydro-2H- pyran-3,4,5-triyl triacetate
A solution of Example 2.57.4 (0.235 g) in tetrahydrofuran (1.0 mL) and methanol (1.0 mL) was cooled to 0 °C, and sodium borohydride (6.07 mg) was added in one portion. The reaction was stirred for 15 minutes and was diluted with ethyl acetate (75 mL) and water (50 mL). The organic layer was separated, washed with brine (50 mL), dried over magnesium sulfate, filtered, and concentrated. The residue was purified by silica gel chromatography, eluting with a gradient of 10% to 70% ethyl acetate/heptanes, to provide the title compound. MS (ESI) m/e 734.5 (M+H)+.
2.57.6 (2S,3R,4S,5S,6S)-2-(3-(4-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)butyl)-4-((((4- nitrophenoxy)carbonyl)oxy)methyl)phenoxy)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
To an ambient solution of Example 2.57.5 (0.148 g) and bis(4-nitrophenyl) carbonate (0.123 g) in N,N-dimethylformamide (1.5 mL) was added N,N-diisopropylethylamine (0.053 mL). After 3 hours, the reaction mixture was concentrated. The residue was purified by silica gel chromatography, eluting with a gradient of 10% to 60% ethyl acetate/hexanes, to provide the title compound. MS (ESI) m/e 899.5 (M+H)+.
2.57.7 3-(1-((3-(2-((((2-(4-aminobutyl)-4-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)-57- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (8-(benzo[d]thiazol-2-ylcarbamoyl)naphthalen-2-yl)picolinic acid
To a solution of Example 1.6.3 (0.101 g) and Example 2.57.6 (0.095 g) in N,N- dimethylformamide (1.0 mL) was added N,N-diisopropylethylamine (0.055 mL), and the reaction was stirred at room temperature for 3 hours. The reaction was quenched with a mixture of 2,2,2- trifluoroacetic acid (0.204 mL), water (1 mL) and N,N-dimethylformamide (1 mL) and was purified by preparatory reverse-phase HPLC on a Gilson 2020 system using a gradient of 5% to 50% acetonitrile water over 30 minutes. The product-containing fractions were lyophilized to provide the title compound. MS (ESI) m/e 1152.7 (M+H)+.
2.57.8 3-(1-((3-(2-((((2-(4-((R)-2-amino-3-sulfopropanamido)butyl)-4- (((2S,3R,4S,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)naphthalen-2- yl)picolinic acid
To a stirred solution of (R)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3-sulfopropanoic acid (0.058 g) and O-(7-azabenzotriazol-1-yl)-N,N,N’,N’-tetramethyluronium hexafluorophosphate (0.054 g,) in N,N-dimethylformamide (0.5 mL) was added N,N-diisopropylethylamine (0.051 mL). After stirring for 5 minutes, the mixture was added to a mixture of Example 2.57.7 (0.113 g) and N,N- diisopropylethylamine (0.051 mL) in N,N-dimethylformamide (0.5 mL). After stirring for 2 hours, diethylamine (0.102 mL) was added, and the reaction mixture was stirred for 30 minutes. The reaction mixture was diluted with a solution of 2,2,2-trifluoroacetic acid (0.189 mL) in water (1 mL) and was purified by preparatory reverse-phase HPLC on a Gilson 2020 system using a gradient of 5% to 85% acetonitrile water over 30 minutes. The product-containing fractions were lyophilized to provide the title compound. MS (ESI) m/e 1303.1 (M+H)+.
2.57.9 4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)naphthalen-2-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-3-[4-({N-[6-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-3-sulfo-L- alanyl}amino)butyl]phenyl beta-D-glucopyranosiduronic acid
To a solution of Example 2.57.8 (0.044 g) and 2,5-dioxopyrrolidin-1-yl 6-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)hexanoate (0.012 g) in N,N-dimethylformamide (0.4 mL) was added N,N- diisopropylethylamine (0.027 mL), and the reaction mixture was stirred for 2 hours at room temperature The reaction mixture was quenched with a mixture of 222-trifluoroacetic acid (0060 mL), water (1 mL) and N,N-dimethylformamide (1 mL) and purified by preparatory reverse-phase HPLC on a Gilson 2020 system using a gradient of 5% to 50% acetonitrile water over 30 minutes. The product-containing fractions were lyophilized to provide the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ 13.10 (s, 1H), 9.02 (s, 1H), 8.38 (dd, 1H), 8.27– 8.14 (m, 3H), 8.07 (d, 1H), 8.02 (d, 1H), 7.94 (d, 1H), 7.82 (dd, 2H), 7.79– 7.66 (m, 2H), 7.53– 7.44 (m, 1H), 7.48 (s, 1H), 7.37 (t, 1H), 7.23 (d, 1H), 6.98 (s, 2H), 6.88 (d, 1H), 6.82 (dd, 1H), 5.04 (d, 1H), 5.00 (s, 2H), 4.29 (q, 2H), 3.57 (s, 2H), 3.44 (s, 4H), 3.41 (d, 1H), 3.40– 3.27 (m, 3H), 3.30– 3.21 (m, 2H), 3.03 (t, 2H), 2.85 (s, 3H), 2.79 (dd, 1H), 2.70 (dd, 1H), 2.58 (s, 2H), 2.23 (s, 3H), 2.06 (t, 2H), 1.53– 1.41 (m, 5H), 1.42 (s, 6H), 1.26 (s, 2H), 1.25– 1.07 (m, 8H), 0.85 (s, 6H). MS (ESI) m/e 1494.1 (M-H)-.
2.58 Synthesis of 2-{6-[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl]- 2-methyl-3,3-dioxido-7-oxo-8-oxa-3lambda6-thia-2,6-diazanonan-9-yl}-5- (4-{[(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetyl]amino}butyl)phenyl beta-D-glucopyranosiduronic acid (Synthon UH)
2.58.1 (9H-fluoren-9-yl)methyl but-3-yn-1-ylcarbamate
A solution of but-3-yn-1-amine hydrochloride (9 g) and N,N-diisopropylethylamine (44.7 mL) was stirred in dichloromethane(70 mL) and the mixture was cooled to 0 °C. A solution of (9H- fluoren-9-yl)methyl carbonochloridate (22.06 g) in dichloromethane (35 mL) was added, and the reaction mixture was stirred for 2 hours. The reaction mixture was concentrated, and the residue was purified by silica gel chromatography, eluting with petroleum ether in ethyl acetate (10%-25%) to provide the title compound. MS (ESI) m/e 314 (M+Na)+.
2.58.2 (2S,3S,4S,5R,6S)-methyl 6-(5-(4-(((9H-fluoren-9- yl)methoxy)carbonylamino)but-1-ynyl)-2-formylphenoxy)- 3,4,5-triacetoxy-tetrahydro-2H-pyran-2-carboxylate
Example 2.58.3 (2.7 g), Example 2.58.1 (2.091 g), bis(triphenylphosphine)palladium(II) chloride (0.336 g) and copper(I) iodide (0.091 g) were weighed into a vial and flushed with a stream of nitrogen. Triethylamine (2.001 mL) and tetrahydrofuran (45 mL) were added, and the reaction was stirred at room temperature. After stirring for 16 hours, the reaction mixture was diluted with ethyl acetate (200 mL) and washed with water (100 mL) and brine (100 mL). The organic layer was dried over magnesium sulfate, filtered, and concentrated. The residue was purified by silica gel chromatography, eluting with petroleum ether in ethyl acetate (10%-50%), to provide the title compound. MS (ESI) m/e 750 (M+Na)+.
2.58.3 (2S,3S,4S,5R,6S)-methyl 6-(5-(4-(((9H-fluoren-9- yl)methoxy)carbonylamino)butyl)-2-formylphenoxy)-3,4,5- triacetoxy-tetrahydro-2H-pyran-2-carboxylate Example 2.58.2 (1.5 g) and tetrahydrofuran (45 mL) were added to 10% Pd-C (0.483 g) in a 100 mL pressure bottle and the mixture was stirred for 16 hours under 1 atm H2 at room temperature. The reaction mixture was filtered and concentrated to provide the title compound. MS (ESI) m/e 754 (M+Na)+.
2.58.4 (2S,3S,4S,5R,6S)-methyl 6-(5-(4-(((9H-fluoren-9- yl)methoxy)carbonylamino)butyl)-2- (hydroxymethyl)phenoxy)-3,4,5-triacetoxy-tetrahydro-2H- pyran-2-carboxylate
A solution of Example 2.58.3 (2.0 g) in tetrahydrofuran (7.00 mL) and methanol (7 mL) was cooled to 0 °C and NaBH4 (0.052 g) was added in one portion. After 30 minutes, the reaction mixture was diluted with ethyl acetate (150 mL) and water (100 mL). The organic layer was separated, washed with brine (100 mL), dried over magnesium suflate, filtered, and concentrated. The residue was purified by silica gel chromatography, eluting with petroleum ether in ethyl acetate (10%-40%), to provide the title compound. MS (ESI) m/e 756 (M+Na)+.
2.58.5 (2S,3S,4S,5R,6S)-methyl 6-(5-(4-(((9H-fluoren-9- yl)methoxy)carbonylamino)butyl)-2-(((4- nitrophenoxy)carbonyloxy)methyl)phenoxy)-3,4,5-triacetoxy- tetrahydro-2H-pyran-2-carboxylate
To a solution of Example 2.58.4 (3.0 g) and bis(4-nitrophenyl) carbonate (2.488 g) in dry acetonitrile (70 mL) at 0 °C was added N,N-diisopropylethylamine (1.07 mL). After stirring at room temperature for 16 hours, the reaction mixture was concentrated to give a residue, which was purified by silica gel chromatography, eluting with petroleum ether in ethyl acetate (10%-50%), to provide the title compound. MS (ESI) m/e 921 (M+Na)+.
2.58.6 3-(1-((3-(2-((((4-(4-aminobutyl)-2-(((2R,3S,4R,5R,6R)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(2-(N,N- dimethylsulfamoyl)ethyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)picolinic acid
To a cold (0 °C) solution of Example 2.58.5 (40.8 mg) and Example 1.36 (40 mg) in N,N- dimethylformamide (4 mL) was added N,N-diisopropylethylamine (0.026 mL). The reaction mixture was slowly warmed to room temperature and stirred overnight. To the reaction mixture was added water (2 mL) and LiOH H2O (50 mg), and the mixture was stirred at room temperature for 3 hours. The mixture was acidified with trifluoroacetic acid, filtered and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1%
trifluoroacetic acid to provide the title compound MS (ESI) m/e 1278.7 (M-H)-.
2.58.7 2-{6-[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl- 1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl]-2-methyl-3,3-dioxido-7-oxo-8-oxa-3lambda6-thia- 2,6-diazanonan-9-yl}-5-(4-{[(2,5-dioxo-2,5-dihydro-1H-pyrrol- 1-yl)acetyl]amino}butyl)phenyl beta-D-glucopyranosiduronic acid
To a solution of Example 2.58.6 (35.1 mg) in N,N-dimethylformamide (4 mL) was added 2,5- dioxopyrrolidin-1-yl 2-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetate (6.93 mg) and N,N- diisopropylethylamine (0.026 mL). The reaction mixture was stirred at room temperature overnight. The mixture was diluted with N,N-dimethylformamide (2 mL), filtered and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1% trifluoroacetic acid, to provide the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 12.85 (s, 1H), 8.02 (dd, 2H), 7.76 (d, 1H), 7.58 (d, 1H), 7.53– 7.37 (m, 3H), 7.32 (td, 2H), 7.24 (s, 1H), 7.16 (dd, 1H), 7.04 (s, 2H), 6.99– 6.87 (m, 2H), 6.81 (d, 1H), 5.08 (d, 2H), 4.99 (d, 1H), 4.92 (s, 2H), 3.95 (s, 2H), 3.86 (q, 3H), 3.47– 3.14 (m, 9H), 2.99 (dt, 4H), 2.72 (s, 3H), 2.60 (s, 3H), 2.06 (s, 3H), 1.49 (p, 2H), 1.41– 1.27 (m, 4H), 1.29– 0.86 (m, 10H), 0.80 (d, 7H). MS (ESI) m/e 1413.4 (M- H)-.
2.59 Synthesis of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]-3-(1-{[3-(2-{({[2-{[(2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2-yl]oxy}-4-(4-{[(2,5- dioxo-2,5-dihydro-1H-pyrrol-1- yl)acetyl]amino}butyl)benzyl]oxy}carbonyl)[3-(dimethylamino)-3- oxopropyl]amino}ethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid (Synthon UI)
2.59.1 3-(1-((3-(2-((((4-(4-aminobutyl)-2-(((2R,3S,4R,5R,6R)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(3-(dimethylamino)-3- oxopropyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)picolinic acid
The title compound was prepared as described in Example 2.58.6, substituting Example 1.36 with Example 1.38. MS (ESI) m/e 1243.7 (M+H)+.
2.59.2 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl]-3-(1-{[3-(2-{({[2-{[(2S 3R 4S 5S 6S)-6-carboxy-345- trihydroxytetrahydro-2H-pyran-2-yl]oxy}-4-(4-{[(2,5-dioxo- 2,5-dihydro-1H-pyrrol-1- yl)acetyl]amino}butyl)benzyl]oxy}carbonyl)[3- (dimethylamino)-3-oxopropyl]amino}ethoxy)-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)pyridine-2-carboxylic acid
The title compound was prepared as described in Example 2.58.7, substituting Example 2.58.6 with Example 2.59.1. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 8.02 (dd, 2H), 7.76 (d, 1H), 7.58 (d, 1H), 7.44 (ddd, 3H), 7.32 (td, 2H), 7.24 (s, 1H), 7.13 (dd, 1H), 7.04 (s, 2H), 6.99– 6.86 (m, 2H), 6.81 (d, 1H), 5.06 (d, 2H), 4.98 (d, 1H), 4.92 (s, 2H), 3.95 (s, 2H), 3.85 (q, 3H), 3.77 (d, 2H), 3.39 (q, 5H), 3.27 (q, 4H), 2.99 (dt, 4H), 2.88 (s, 2H), 2.81– 2.66 (m, 5H), 2.06 (d, 3H), 1.50 (p, 2H), 1.34 (dd, 4H), 1.27– 0.85 (m, 9H), 0.79 (d, 6H). MS (ESI) m/e 1401.3 (M+H)+.
2.60 Synthesis of 2-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-sulfamoylethyl)carbamoyl}oxy)methyl]-5-(4-{[(2,5-dioxo- 2,5-dihydro-1H-pyrrol-1-yl)acetyl]amino}butyl)phenyl beta-D- glucopyranosiduronic acid (Synthon US)
2.60.1 3-(1-((3-(2-((((4-(4-aminobutyl)-2-(((2R,3S,4R,5R,6R)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(2-sulfamoylethyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl)picolinic acid
The title compound was prepared as described in Example 2.58.6, substituting Example 1.36 with Example 1.18.20. MS (ESI) m/e 1251.2 (M+H)+.
2.60.2 2-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl- 1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-sulfamoylethyl)carbamoyl}oxy)methyl]-5-(4- {[(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)acetyl]amino}butyl)phenyl beta-D-glucopyranosiduronic acid
The title compound was prepared as described in Example 2.58.7, substituting Example 2.58.6 with Example 2.60.1. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 12.84 (s, 2H), 8.04 (dd, 2H), 7.77 (d, 1H), 7.60 (d, 1H), 7.53– 7.38 (m, 3H), 7.38– 7.30 (m, 2H), 7.26 (s, 1H), 7.16 (d, 1H) 705 (s 2H) 696– 677 (m 5H) 509 (s 2H) 500 (d 1H) 494 (s 2H) 397 (s 2H) 387 (q 3H), 3.48– 3.16 (m, 5H), 3.09– 2.94 (m, 4H), 2.07 (s, 3H), 1.50 (d, 2H), 1.36 (d, 3H), 1.29– 0.88 (m, 9H), 0.81 (d, 7H). MS (ESI) m/e 1385.5 (M-H)-.
2.61 Synthesis of 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]-3-(1-{[3-(2-{({[2-{[(2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2-yl]oxy}-4-(4-{[(2,5- dioxo-2,5-dihydro-1H-pyrrol-1- yl)acetyl]amino}butyl)benzyl]oxy}carbonyl)[3-(methylamino)-3- oxopropyl]amino}ethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid (Synthon UY)
2.61.1 3-(1-((3-(2-((((4-(4-aminobutyl)-2-(((2R,3S,4R,5R,6R)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(3-(methylamino)-3- oxopropyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)picolinic acid
The title compound was prepared as described in Example 2.58.6, substituting Example 1.36 with Example 1.39. MS (ESI) m/e 1228.8 (M+H)+.
2.61.2 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl]-3-(1-{[3-(2-{({[2-{[(2S,3R,4S,5S,6S)-6-carboxy-3,4,5- trihydroxytetrahydro-2H-pyran-2-yl]oxy}-4-(4-{[(2,5-dioxo- 2,5-dihydro-1H-pyrrol-1- yl)acetyl]amino}butyl)benzyl]oxy}carbonyl)[3-(methylamino)- 3-oxopropyl]amino}ethoxy)-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)pyridine-2-carboxylic acid
The title compound was prepared as described in Example 2.58.7, substituting Example 2.58.6 with Example 2.61.1. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ ppm 12.83 (s, 1H), 8.06 (s, 1H), 8.01 (dd, 1H), 7.77 (d, 1H), 7.71 (d, 0H), 7.60 (d, 1H), 7.45 (tdd, 3H), 7.38– 7.29 (m, 2H), 7.26 (s, 1H), 7.15 (d, 1H), 7.05 (d, 1H), 6.96– 6.90 (m, 2H), 6.82 (d, 1H), 5.07 (s, 2H), 5.01 (t, 1H), 4.94 (s, 2H), 3.97 (s, 2H), 3.87 (q, 3H), 3.79 (d, 2H), 3.28 (p, 2H), 3.09– 2.93 (m, 3H), 2.52 (d, 3H), 2.35– 2.26 (m, 2H), 2.07 (d, 2H), 1.60– 1.44 (m, 2H), 1.34 (d, 3H), 1.29– 0.88 (m, 6H), 0.81 (d, 5H). MS (ESI) m/e 1363.5 (M-H)-.
2.62 Synthesis of 3-{1-[(3-{2-[(3-amino-3-oxopropyl)({[2-{[(2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2-yl]oxy}-4-(4-{[(2,5- dioxo-2,5-dihydro-1H-pyrrol-1- yl)acetyl]amino}butyl)benzyl]oxy}carbonyl)amino]ethoxy}-57- dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol-4-yl}- 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)- yl]pyridine-2-carboxylic acid (Synthon UX)
2.62.1 3-(1-((3-(2-((3-amino-3-oxopropyl)(((4-(4-aminobutyl)-2- (((2R,3S,4R,5R,6R)-6-carboxy-3,4,5-trihydroxytetrahydro-2H- pyran-2-yl)oxy)benzyl)oxy)carbonyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)picolinic acid
The title compound was prepared as described in Example 2.58.6, substituting Example 1.36 with Example 1.32.2. MS (ESI) m/e 1214.6 (M+H)+.
2.62.2 3-{1-[(3-{2-[(3-amino-3-oxopropyl)({[2-{[(2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2-yl]oxy}-4-(4- {[(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)acetyl]amino}butyl)benzyl]oxy}carbonyl)amino]ethoxy}-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H- pyrazol-4-yl}-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]pyridine-2-carboxylic acid
The title compound was prepared as described in Example 2.58.7, substituting Example 2.58.6 with Example 2.62.1. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ ppm 12.83 (s, 2H), 8.06 (s, 1H), 8.01 (d, 1H), 7.77 (d, 1H), 7.60 (d, 1H), 7.53– 7.38 (m, 3H), 7.34 (q, 2H), 7.26 (s, 1H), 7.15 (d, 1H), 7.05 (s, 2H), 6.93 (d, 2H), 6.87– 6.73 (m, 2H), 5.07 (d, 2H), 5.04– 4.97 (m, 1H), 4.94 (s, 2H), 3.97 (s, 2H), 3.87 (q, 3H), 3.79 (d, 2H), 3.29 (t, 3H), 3.10– 2.95 (m, 4H), 2.32 (p, 2H), 2.07 (d, 3H), 1.51 (dd, 2H), 1.36 (dd, 5H), 1.30– 0.86 (m, 8H), 0.81 (d, 6H). MS (ESI) m/e 1349.5 (M-H)-.
2.63 Synthesis of 2-[({[2-({3-[(4-{6-[3-(1,3-benzothiazol-2-ylcarbamoyl)-1H- indol-5-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]- 5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-5-(4-{[(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)acetyl]amino}butyl)phenyl beta-D- glucopyranosiduronic acid (Synthon WZ)
2.63.1 3-(1-((3-(2-((((4-(4-aminobutyl)-2-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(methyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (3-(benzo[d]thiazol-2-ylcarbamoyl)-1H-indol-5-yl)picolinic acid
The title compound was prepared by substituting Example 1.34.5 for Example 1.12.10 and Example 2585 for Example 2116 in Example 2117 2.63.2 2-[({[2-({3-[(4-{6-[3-(1,3-benzothiazol-2-ylcarbamoyl)-1H- indol-5-yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1- yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](methyl)carbamoyl}oxy)methyl]-5-(4-{[(2,5-dioxo- 2,5-dihydro-1H-pyrrol-1-yl)acetyl]amino}butyl)phenyl beta-D- glucopyranosiduronic acid
The title compound was prepared by substituting Example 2.63.1 for Example 2.9.1 in Example 2.10. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 12.47 (bs, 1H), 12.16 (d, 1H), 9.01 (s, 1H), 8.69 (d, 1H), 8.11-8.04 (m, 4H), 7.99 (d, 1H), 7.76 (d, 1H), 7.64 (d, 1H), 7.48 (s, 1H), 7.45 (t, 1H), 7.31 (t, 1H), 7.19 (t, 1H), 7.07 (s, 1H), 6.94 (s, 1H), 6.86 (d, 1H), 5.10 (s, 2H), 5.03 (d, 1H), 3.99 (s, 2H), 3.90 (m, 3H), 3.48 (m, 3H), 3.28 (m, 2H), 3.05 (m, 4H), 2.93 (s, 2H), 2.88 (s, 2H), 2.54-2.53 (m, 2H), 2.24 (s, 3H), 1.54 (m, 2H), 1.40 (m, 4H), 1.30-1.22 (m, 6H), 1.20-1.14 (m, 6H), 1.11-0.96 (m, 2H), 0.87 (d, 6H). MS (ESI) m/e 1300 (M+Na)+, 1276 (M-H)-.
2.64 Synthesis of 2-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-5,6- dihydroimidazo[1,5-a]pyrazin-7(8H)-yl]-2-carboxypyridin-3-yl}-5- methyl-1H-pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl]carbamoyl}oxy)methyl]-5-(4-{[(2,5-dioxo-2,5-dihydro-1H- pyrrol-1-yl)acetyl]amino}butyl)phenyl beta-D-glucopyranosiduronic acid (Synthon XO)
2.64.1 3-(1-((3-(2-((((4-(4-aminobutyl)-2-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (1-(benzo[d]thiazol-2-ylcarbamoyl)-5,6-dihydroimidazo[1,5- a]pyrazin-7(8H)-yl)picolinic acid
The title compound was prepared by substituting Example 1.4.10 for Example 1.12.10 and Example 2.58.5 for Example 2.11.6 in Example 2.11.7. MS (ESI) m/e 1133 (M+H)+, 1131 (M-H)-.
2.64.2 2-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-5,6- dihydroimidazo[1,5-a]pyrazin-7(8H)-yl]-2-carboxypyridin-3- yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl]carbamoyl}oxy)methyl]-5-(4-{[(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)acetyl]amino}butyl)phenyl beta-D- glucopyranosiduronic acid
The title compound was prepared by substituting Example 2.64.1 for Example 2.9.1 in Example 2.10. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 8.08 (t, 1H), 8.01 (s, 1H), 7.99 (d, 1H) 776 (d 1H) 761 (d 1H) 746 (t 1H) 734 (s 1H) 733 (t 1H) 717 (m 3H) 708 (s 2H) 6.92 (s, 1H), 6.84 (d, 1H), 5.12 (s, 2H), 5.05 (s, 2H), 5.02 (d, 1H), 4.27 (m, 2H), 4.10 (m, 2H), 3.99 (s, 2H), 3.91 (m, 2H), 3.84 (s, 2H), 3.70 (m, 2H), 3.42 (t, 2H), 3.35 (t, 2H), 3.30 (t, 2H), 3.06 (m, 5H), 2.53 (m, 2H), 2.14 (s, 3H), 1.53 (m, 2H), 1.43-1.35 (m, 4H), 1.27 (m, 4H), 1.14 (q, 4H), 1.03 (dd, 2H), 0.86 (s, 6H). MS (ESI) m/e 1270 (M+H)+, 1268 (M-H)-.
2.65 Synthesis of (6S)-2,6-anhydro-6-(2-{2-[({[2-({3-[(4-{6-[1-(1,3- benzothiazol-2-ylcarbamoyl)-5,6-dihydroimidazo[1,5-a]pyrazin-7(8H)- yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl]carbamoyl}oxy)methyl]-5- ({N-[(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetyl]-L-valyl-L- alanyl}amino)phenyl}ethyl)-L-gulonic acid (Synthon XW)
2.65.1 (3R,4S,5R,6R)-3,4,5-tris(benzyloxy)-6-(benzyloxymethyl)- tetrahydropyran-2-one
To a solution of (3R,4S,5R,6R)-3,4,5-tris(benzyloxy)-6-((benzyloxy)methyl)tetrahydro-2H- pyran-2-ol (75 g) in dimethyl sulfoxide (400 mL) at 0 °C was added acetic anhydride (225 mL). The mixture was stirred for 16 hours at room temperature before cooled to 0 °C. A large volume of water was added, and the stirring was stopped and the reaction mixture was allowed to settle for 3 hours (the crude lactone was at the bottom of the flask). The supernatant was removed, and the crude mixture was diluted with ethyl acetate, washed 3 times with water, neutralized with saturated aqueous solution of NaHCO3, and washed again twice with water. The organic layer was then dried over magnesium sulfate, filtered and concentrated to provide the title compound. MS (ESI) m/e 561 (M+Na)+.
2.65.2 (3R,4S,5R,6R)-3,4,5-tris(benzyloxy)-6-(benzyloxymethyl)-2- ethynyl- tetrahydro-2H-pyran-2-ol
To a solution of ethynyltrimethylsilane (18.23 g) in tetrahydrofuran (400 mL) under nitrogen and chilled in a dry ice/acetone bath (internal temp -65oC) was added 2.5M BuLi in hexane (55.7 mL) dropwise, keeping the temperature below -60 °C. The mixture was stirred in a cold bath for 40 minutes, followed by an ice-water bath (internal temp rose to 0.4 °C) for 40 minutes, and finally cooled to -75 °C again. A solution of Example 2.55.1 (50 g) in tetrahydrofuran (50 mL) was added dropwise, keeping the internal temperature below -70 °C. The mixture was stirred in a dry ice/acetone bath for an additional 3 hours. The reaction mixture was quenched with saturated aqueous NaHCO3 solution (250 mL). The mixture was allowed to warm to room temperature, extracted with ethyl acetate (3x 300 mL), dried over MgSO4, filtered, and concentrated in vacuo to provide the title compound. MS (ESI) m/e 659 (M+Na)+.
2.65.3 trimethyl(((3S,4R,5R,6R)-3,4,5-tris(benzyloxy)-6- (benzyloxymethyl)-tetrahydro-2H-pyran-2-yl)ethynyl)silane
To a mixture of Example 2.65.2 (60 g) in acetonitrile (450 mL) and dichloromethane(150 mL) at -15 oC in an ice-salt bath was added triethylsilane (81 mL) dropwise, followed by addition of boron trifluoride diethyl ether complex (406 mL) at such a rate that the internal temperature did not exceed -10 °C. The mixture was stirred between -15 oC and -10 oC for 2 hours. The reaction mixture was quenched with saturated aqueous NaHCO3 solution (275 mL) and stirred for 1 hour at room temperature. The mixture was extracted with ethyl acetate (3 x 550 mL). The combined extracts were dried over MgSO4, filtered, and concentrated. The residue was purified by flash
chromatography eluting with a gradient of 0% to 7% ethyl acetate/petroleum ether to provide the title compound. MS (ESI) m/e 643 (M+Na)+.
2.65.4 (2R,3R,4R,5S)-3,4,5-tris(benzyloxy)-2-(benzyloxymethyl)-6- ethynyl-tetrahydro-2H-pyran
To a mixed solution of Example 2.65.3 (80 g) in dichloromethane(200 mL) and methanol (1000 mL) was added 1N aqueous NaOH solution (258 mL). The mixture was stirred at room temperature for 2 hours. The solvent was removed. The residue was then partitioned between water and dichloromethane. The extracts were washed with brine, dried over Na2SO4, filtered, and concentrated to provide the title compound. MS (ESI) m/e 571 (M+Na)+.
2.65.5 (2R,3R,4R,5S)-2-(acetoxymethyl)-6-ethynyl-tetrahydro-2H- pyran-3,4,5-triyl triacetate
To a solution of Example 2.65.4 (66 g) in acetic anhydride (500 mL) cooled by an ice/water bath was added boron trifluoride diethyl ether complex (152 mL) dropwise. The mixture was stirred at room temperature for 16 hours, cooled with an ice/water bath and neutralized with saturated aqueous NaHCO3 solution. The mixture was extracted with ethyl acetate (3x500 mL), dried over Na2SO4 and concentrated in vacuo. The residue was purified by flash chromatography eluting with a gradient of 0% to 30% ethyl acetate/petroleum ether to provide the title compound. MS (ESI) m/e 357 (M+H)+.
2.65.6 (3R,4R,5S,6R)-2-ethynyl-6-(hydroxymethyl)-tetrahydro-2H- pyran-3,4,5-triol
To a solution of Example 2.65.5 (25 g) in methanol (440 mL) was added sodium methanolate (2.1 g). The mixture was stirred at room temperature for 2 hours, then neutralized with 4M HCl in dioxane. The solvent was removed, and the residue was adsorbed onto silica gel and loaded onto a silica gel column. The column was eluted with a gradient of 0 to 100% ethyl acetate/petroleum ether then 0% to 12% methanol/ethyl acetate to provide the title compound. MS (ESI) m/e 211 (M+Na)+.
2.65.7 (2S,3S,4R,5R)-6-ethynyl-3,4,5-trihydroxy-tetrahydro-2H- pyran-2-carboxylic acid
A three-necked round bottom flask was charged with Example 2.65.6 (6.00 g), KBr (0.30 g), tetrabutylammonium bromide (0.41 g) and 60 mL of saturated aqueous NaHCO3 solution. (2,2,6,6- Tetramethylpiperidin-1-yl)oxidanyl (0.15 g) in 60 mL dichloromethane was added. The mixture was stirred vigorously and cooled in an ice-salt bath to -2 °C internal temperature. A solution of brine (12 mL), aqueous NaHCO3 solution (24 mL) and NaOCl (154 mL) was added dropwise such that the internal temperature was maintained below 2 °C. The pH of the reaction mixture was maintained in the 82-84 range with the addition of solid Na2CO3 After a total of 6 hours the reaction was cooled to 3 °C internal temperature and ethanol (~20 mL) was added dropwise and was stirred for ~ 30 minutes. The mixture was transferred to a separatory funnel, and the dichloromethane layer was discarded. The pH of the aqueous layer was adjusted to 2-3 using 1 M aqueous HCl. The aqueous layer was then concentrated to dryness. Methanol (100 mL was) added to the dry solid, and the slurry was stirred for ~30 minutes. The mixture was filtered over a pad of diatomaceous earth, and the residue in the funnel was washed with ~100 mL of methanol. The filtrate was concentrated under reduced pressure to obtain the title compound.
2.65.8 (2S,3S,4R,5R)-methyl 6-ethynyl-3,4,5-trihydroxytetrahydro- 2H-pyran-2-carboxylate
A 500 mL three-necked round bottom flask was charged with a suspension of Example 2.65.7 (6.45 g) in methanol (96 mL) and was cooled in an ice-salt-bath with internal temperature of -1 °C. Neat thionyl chloride (2.79 mL) was carefully added. The internal temperature kept rising throughout the addition but did not exceed 10 °C. The reaction was allowed to slowly warm up to 15-20 °C over 2.5 hours. After 2.5 hours, the reaction was concentrated to provide the title compound.
2.65.9 (3S,4R,5S,6S)-2-ethynyl-6-(methoxycarbonyl)tetrahydro-2H- pyran-3,4,5-triyl triacetate
Example 2.65.8 (6.9 g) as a solution in N,N-dimethylformamide (75 mL) was added 4- dimethylaminopyridine (0.17 g) and acetic anhydride (36.1 mL). The suspension was cooled in an ice-bath and pyridine (18.04 mL) was added via syringe over 15 minutes. The reaction was allowed to warm to room temperature overnight. Additional acetic anhydride (12 mL) and pyridine (6 mL) were added and stirring was continued for an additional 6 hours. The reaction was cooled in an ice- bath and 250 mL of saturated aqueous NaHCO3 solution was added and stirred for 1 hour. Water (100 mL) was added, and the mixture was extracted with ethyl acetate. The organic extract was washed twice with saturated CuSO4 solution, dried and concentrated. The residue was purified by flash chromatography, eluting with 50% ethyl acetate/petroleum ether to provide the title compound. 1H NMR (500 MHz, methanol-d4) δ ppm 5.29 (t, 1H), 5.08 (td, 2H), 4.48 (dd, 1H), 4.23 (d, 1H), 3.71 (s, 3H), 3.04 (d, 1H), 2.03 (s, 3H), 1.99 (s, 3H), 1.98 (s, 4H). MS (ESI) m/e 359.9 (M+NH4)+.
2.65.10 2-iodo-4-nitrobenzoic acid
A 3L fully jacketed flask equipped with a mechanical stirrer, temperature probe and an addition funnel under a nitrogen atmosphere, was charged with 2-amino-4-nitrobenzoic acid (69.1 g, Combi-Blocks) and sulfuric acid, 1.5 M aqueous (696 mL). The resulting suspension was cooled to 0 °C internal temperature, and a solution of sodium nitrite (28.8 g) in water (250 mL) was added dropwise over 43 minutes with the temperature kept below 1 °C. The reaction mixture was stirred at ca.0 °C for 1 hour. A solution of potassium iodide (107 g) in water (250 mL) was added dropwise over 44 minutes with the internal temperature kept below 1 °C. (Initially addition was exothermic and there was gas evolution). The reaction mixture was stirred 1 hour at 0 °C. The temperature was raised to 20 °C and then stirred at ambient temperature overnight. The reaction mixture became a suspension. The reaction mixture was filtered, and the collected solid was washed with water. The wet solid (~ 108 g) was stirred in 10 % sodium sulfite (350 mL, with ~ 200 mL water used to wash in the solid) for 30 minutes. The suspension was acidified with concentrated hydrochloric acid (35 mL), and the solid was collected by filtration and washed with water. The solid was slurried in water (1L) and re-filtered, and the solid was left to dry in the funnel overnight. The solid was then dried in a vacuum oven for 2 hours at 60 °C. The resulting solid was triturated with dichloromethane (500 mL), and the suspension was filtered and washed with additional dichloromethane. The solid was air-dried to provide the title compound. MS (ESI) m/e 291.8 (M-H)-.
2.65.11 (2-iodo-4-nitrophenyl)methanol
A flame-dried 3 L 3-necked flask was charged with Example 2.65.10 (51.9 g) and
tetrahydrofuran (700 mL). The solution was cooled in an ice bath to 0.5 °C, and borane- tetrahydrofuran complex (443 mL, 1M in THF) was added dropwise (gas evolution) over 50 minutes, reaching a final internal temperature of 1.3 °C. The reaction mixture was stirred for 15 minutes, and the ice bath was removed. The reaction left to come to ambient temperature over 30 minutes. A heating mantle was installed, and the reaction was heated to an internal temperature of 65.5 °C for 3 hours, and then allowed to cool to room temperature while stirring overnight. The reaction mixture was cooled in an ice bath to 0 °C and quenched by dropwise addition of methanol (400 mL). After a brief incubation period, the temperature rose quickly to 2.5 °C with gas evolution. After the first 100 mL are added over ~ 30 minutes, the addition was no longer exothermic, and the gas evolution ceased. The ice bath was removed, and the mixture was stirred at ambient temperature under nitrogen overnight. The mixture was concentrated to a solid, dissolved in dichloromethane/methanol and adsorbed on to silica gel (~ 150 g). The residue was loaded on a plug of silica gel (3000 mL) and eluted with dichloromethane to provide the title compound. MS (DCI) m/e 296.8 (M+NH4)+.
2.65.12 (4-amino-2-iodophenyl)methanol
A 5 L flask equipped with a mechanical stirrer, heating mantle controlled by a JKEM temperature probe and condenser was charged with Example 2.65.11 (98.83 g) and ethanol (2 L). The reaction was stirred rapidly, and iron (99 g) was added, followed by a solution of ammonium chloride (20.84 g) in water (500 mL). The reaction was heated over the course of 20 minutes to an internal temperature of 80.3 °C, when it began to reflux vigorously. The mantle was dropped until the reflux calmed. Thereafter, the mixture was heated to 80 °C for 1.5 hour. The reaction was filtered hot through a membrane filter, and the iron residue was washed with hot 50% ethyl acetate/methanol (800 mL). The eluent was passed through a diatomaceous earth pad, and the filtrate was concentrated. The residue was partitioned between 50% brine (1500 mL) and ethyl acetate (1500 mL). The layers were separated, and the aqueous layer was extracted with ethyl acetate (400 mL x 3). The combined organic layers were dried over sodium sulfate, filtered and concentrated to provide the title compound which was used without further purification MS (DCI) m/e 2669 (M+NH4)+ 2.65.13 4-(((tert-butyldimethylsilyl)oxy)methyl)-3-iodoaniline
A 5 L flask with a mechanical stirrer was charged with Example 2.65.12 (88 g) and dichloromethane (2 L). The suspension was cooled in an ice bath to an internal temperature of 2.5 °C, and tert-butylchlorodimethylsilane (53.3 g) was added portion-wise over 8 minutes. After 10 minutes, 1H-imidazole (33.7 g) was added portionwise to the cold reaction. The reaction was stirred 90 minutes while the internal temperature rose to 15 °C. The reaction mixture was diluted with water (3 L) and dichloromethane (1 L). The layers were separated, and the organic layer was dried over sodium sulfate, filtered, and concentrated to an oil. The residue was purified by silica gel chromatography (1600 g silica gel), eluting a gradient of 0 - 25% ethyl acetate in heptane, to provide the title compound.
2.65.14 (S)-2-((S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3- methylbutanamido)propanoic acid
To a solution of (S)-2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3-methylbutanoic acid (6.5 g) in dimethoxyethane (40 mL) was added (S)-2-aminopropanoic acid (1.393 g) and sodium bicarbonate (1.314 g) in water (40 mL). Tetrahydrofuran (20 mL) was added to aid solubility. The resulting mixture was stirred at room temperature for 16 hours. Aqueous citric acid (15%, 75 mL) was added, and the mixture was extracted with 10% 2-propanol in ethyl acetate (2 x 100 mL). A precipitate formed in the organic layer. The combined organic layers were washed with water (2 x 150 mL). The organic layer was concentrated under reduced pressure and then triturated with diethyl ether (80 mL). After brief sonication, the title compound was collected by filtration. MS (ESI) m/e 411 (M+H)+.
2.65.15 (9H-fluoren-9-yl)methyl ((S)-1-(((S)-1-((4-(((tert- butyldimethylsilyl)oxy)methyl)-3-iodophenyl)amino)-1- oxopropan-2-yl)amino)-3-methyl-1-oxobutan-2-yl)carbamate
A solution of Example 2.65.13 (5.44 g) and Example 2.65.14 (6.15 g) in a mixture of dichloromethane (70 mL) and methanol (35.0 mL) was added ethyl 2-ethoxyquinoline-1(2H)- carboxylate (4.08 g), and the reaction was stirred overnight. The reaction mixture was concentrated and the residue was loaded onto silica gel, eluting with a gradient of 10% to 95% heptane in ethyl acetate followed by 5% methanol in dichloromethane. The product-containing fractions were concentrated, dissolved in 0.2% methanol in dichloromethane (50 mL), loaded onto silica gel and eluted with a gradient of 0.2% to 2% methanol in dichloromethane. The product containing fractions were collected to provide the title compound. MS (ESI) m/e 756.0 (M+H)+.
2.65.16 (2S,3S,4R,5S,6S)-2-((5-((S)-2-((S)-2-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)-3- methylbutanamido)propanamido)-2-(((tert- butyldimethylsilyl)oxy)methyl)phenyl)ethynyl)-6- (methoxycarbonyl)tetrahydro-2H-pyran-345-triyl triacetate A solution of Example 2.65.9 (4.500 g), Example 2.65.15 (6.62 g), copper(I) iodide (0.083 g) and bis(triphenylphosphsine)palladium(II) dichloride (0.308 g) were combined in vial and degassed. N,N-dimethylformamide (45 mL) and N-ethyl-N-isopropylpropan-2-amine (4.55 mL) were added, and the reaction vessel was flushed with nitrogen and stirred at room temperature overnight. The reaction was partitioned between water (100 mL) and ethyl acetate (250 mL). The layers were separated, and the organic layer was dried over magnesium sulfate, filtered, and concentrated. The residue was purified by silica gel chromatography, eluting with a gradient of 5% to 95% ethyl acetate in heptane. The product containing fractions were collected, concentrated and purified by silica gel chromatography, eluting with a gradient of 0.25% to 2.5% methanol in dichloromethane to provide the title compound. MS (ESI) m/e 970.4 (M+H)+.
2.65.17 (2S,3S,4R,5S,6S)-2-(5-((S)-2-((S)-2-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)-3- methylbutanamido)propanamido)-2-(((tert- butyldimethylsilyl)oxy)methyl)phenethyl)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
Example 2.65.16 (4.7 g) and tetrahydrofuran (95 mL) were added to 5% Pt/C (2.42 g, wet) in a 50 mL pressure bottle and the reaction was shaken for 90 minutes at room temperature under 50 psi of hydrogen. The reaction mixture was filtered and concentrated to provide the title compound. MS (ESI) m/e 974.6 (M+H)+.
2.65.18 (2S,3S,4R,5S,6S)-2-(5-((S)-2-((S)-2-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)-3- methylbutanamido)propanamido)-2- (hydroxymethyl)phenethyl)-6-(methoxycarbonyl)tetrahydro- 2H-pyran-3,4,5-triyl triacetate
A solution of Example 2.65.17 (5.4 g) in tetrahydrofuran (7 mL), water (7 mL) and glacial acetic acid (21 mL) was stirred overnight at room temperature. The reaction mixture was diluted with ethyl acetate (200 mL) and was washed with water (100 mL), saturated aqueous NaHCO3 solution (100 mL), and brine (100 mL), dried over magnesium sulfate, filtered, and concentrated. The residue was purified by silica gel chromatography, eluting with a gradient of 0.5% to 5% methanol in dichloromethane, to provide the title compound. MS (ESI) m/e 860.4 (M+H)+.
2.65.19 (2S,3S,4R,5S,6S)-2-(5-((S)-2-((S)-2-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)-3- methylbutanamido)propanamido)-2-((((4- nitrophenoxy)carbonyl)oxy)methyl)phenethyl)-6- (methoxycarbonyl)tetrahydro-2H-pyran-3,4,5-triyl triacetate
To a solution of Example 2.65.18 (4.00 g) and bis(4-nitrophenyl) carbonate (2.83 g) in acetonitrile (80 mL) was added N-ethyl-N-isopropylpropan-2-amine (122 mL) at room temperature After stirring overnight, the reaction mixture was concentrated, dissolved in dichloromethane (250 mL) and washed with saturated aqueous NaHCO3 solution (4 x 150 mL). The organic layer was dried over magnesium sulfate, filtered, and concentrated. The resulting foam was purified by silica gel chromatography, eluting with a gradient of 5% to 75% ethyl acetate in hexanes to provide the title compound. MS (ESI) m/e 1025.5 (M+H)+.
2.65.20 3-(1-((3-(2-((((4-((S)-2-((S)-2-amino-3- methylbutanamido)propanamido)-2-(2-((2S,3R,4R,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)ethyl)benzyl)oxy)carbonyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (1-(benzo[d]thiazol-2-ylcarbamoyl)-5,6-dihydroimidazo[1,5- a]pyrazin-7(8H)-yl)picolinic acid
The title compound was prepared by substituting Example 1.4.10 for Example 1.12.10 and Example 2.65.19 for Example 2.11.6 in Example 2.11.7. MS (ESI) m/e 1257 (M-H)-.
2.65.21 (6S)-2,6-anhydro-6-(2-{2-[({[2-({3-[(4-{6-[1-(1,3-benzothiazol-2- ylcarbamoyl)-5,6-dihydroimidazo[1,5-a]pyrazin-7(8H)-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl]carbamoyl}oxy)methyl]-5-({N-[(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)acetyl]-L-valyl-L- alanyl}amino)phenyl}ethyl)-L-gulonic acid
The title compound was prepared by substituting Example 2.65.20 for Example 2.9.1 in Example 2.10. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 9.88 (s, 1H), 8.26 (t, 2H), 8.00 (m, 2H), 7.76 (d, 1H), 7.61 (d, 1H), 7.46 (m, 2H), 7.38-7.30 (m, 3H), 7.21 (d, 1H), 7.15 (d, 1H), 7.07 (s, 2H), 7.04 (t, 1H), 5.12 (s, 2H), 4.97 (s, 2H), 4.39 (m, 1H), 4.28 (m, 2H), 4.22 (m, 2H), 4.12 (s, 2H), 4.09 (m, 2H), 3.84 (s, 2H), 3.58 (m, 4H), 3.33 (m, 4H), 3.18-3.00 (m, 4H), 2.94 (t, 2H), 2.80-2.55 (m, 2H), 2.13 (s, 3H), 2.08-1.91 (m, 2H), 1.56 (m, 1H), 1.39 (s, 2H), 1.30-1.20 (m, 6H), 1.26-0.95 (m, 6H), 0.85 (m, 12 H). MS (ESI) m/e 1395 (M-H)-.
2.66 Synthesis of (6S)-2,6-anhydro-6-[2-(2-[({[2-({3-[(4-{6-[8-(1,3- benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)- yl]-2-carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2- methoxyethyl)carbamoyl}oxy)methyl]-5-{[N-({(3S,5S)-3-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)-2-oxo-5-[(2-sulfoethoxy)methyl]pyrrolidin-1- yl}acetyl)-L-valyl-L-alanyl]amino}phenyl)ethyl]-L-gulonic acid (Synthon YG)
2661 (3R 7aS)-3-phenyltetrahydropyrrolo[12-c]oxazol-5(3H)-one A solution of (S)-5-(hydroxymethyl)pyrrolidin-2-one (25g), benzaldehyde (25.5g) and para- toluensulfonic acid monohydrate (0.50 g) in toluene (300 mL) was heated to reflux using a Dean- Stark trap under a drying tube for 16 hours. The reaction was cooled to room temperature, and the solvent was decanted from the insoluble materials. The organic layer was washed with saturated aqueous sodium bicarbonate solution (2x) and brine (1x). The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel, eluting with 35/65 heptane/ethyl acetate, to provide the title compound. MS (DCI) m/e 204.0 (M+H)+.
2.66.2 (3R,6R,7aS)-6-bromo-3-phenyltetrahydropyrrolo[1,2-c]oxazol- 5(3H)-one
To a cold (-77 °C) solution of Example 2.66.1 (44.6 g) in tetrahydrofuran (670 mL) was added lithium bis(trimethylsilyl)amide(1.0M in hexanes) (250 mL) dropwise over 40 minutes, keeping Trxn < -73 °C. The reaction mixture was stirred at -77 °C for 2 hours, and bromine (12.5 mL) was added dropwise over 20 minutes, keeping Trxn < -64 °C. The reaction mixture was stirred at -77 °C for 75 minutes and was quenched by the addition of 150 mL cold 10% aqueous sodium thiosulfate solution to the -77 °C reaction. The reaction mixture was warmed to room temperature and partitioned between half-saturated aqueous ammonium chloride solution and ethyl acetate. The layers were separated, and the organic was washed with water and brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel
chromatography, eluting with a gradient of 80/20, 75/25, and 70/30 heptane/ethyl acetate to provide the title compound. MS (DCI) m/e 299.0 and 301.0 (M+NH3+H)+.
2.66.3 (3R,6S,7aS)-6-bromo-3-phenyltetrahydropyrrolo[1,2-c]oxazol- 5(3H)-one
The title compound was isolated as a by-product during the synthesis of Example 2.66.2. MS (DCI) m/e 299.0 and 301.0 (M+NH3+H)+.
2.66.4 (3R,6S,7aS)-6-azido-3-phenyltetrahydropyrrolo[1,2-c]oxazol- 5(3H)-one
To a solution of Example 2.66.2 (19.3 g) in N,N-dimethylformamide (100 mL) was added sodium azide (13.5 g). The reaction mixture was heated to 60 °C for 2.5 hours. The reaction mixture was cooled to room temperature and quenched by the addition of water (500 mL) and ethyl acetate (200 mL). The layers were separated, and the organic layer was washed brine. The combined aqueous layers were back-extracted with ethyl acetate (50 mL). The combined organic layers were dried with sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with 78/22 heptane/ethyl acetate, to provide the title compound. MS (DCI) m/e 262.0 (M+NH3+H)+. 2.66.5 (3R,6S,7aS)-6-amino-3-phenyltetrahydropyrrolo[1,2-c]oxazol- 5(3H)-one
To a solution of Example 2.66.4 (13.5 g) in tetrahydrofuran (500 mL) and water (50 mL) was added polymer-supported triphenylphosphine (55 g). The reaction was mechanically stirred overnight at room temperature. The reaction mixture was filtered through diatomaceous earth, eluting with ethyl acetate and toluene. The solution was concentrated under reduced pressure, dissolved in dichloromethane (100 mL), dried with sodium sulfate, then filtered and concentrated to provide the title compound, which was used in the subsequent step without further purification. MS (DCI) m/e 219.0 (M+H)+.
2.66.6 (3R,6S,7aS)-6-(dibenzylamino)-3-phenyltetrahydropyrrolo[1,2- c]oxazol-5(3H)-one
To a solution of Example 2.66.5 (11.3 g) in N,N-dimethylformamide (100 mL) was added potassium carbonate (7.0 g), potassium iodide (4.2 g), and benzyl bromide (14.5 mL). The reaction was stirred at room temperature overnight and quenched by the addition of water and ethyl acetate. The layers were separated, and the organic layer was washed brine. The combined aqueous layers were back-extracted with ethyl acetate. The combined organic layers were dried with sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel
chromatography, eluting with a gradient of 10 to 15% ethyl acetate in heptane to give a solid that was triturated with heptane to provide the title compound. MS (DCI) m/e 399.1 (M+H)+.
2.66.7 (3S,5S)-3-(dibenzylamino)-5-(hydroxymethyl)pyrrolidin-2-one
To a solution of Example 2.66.6 (13 g) in tetrahydrofuran (130 mL) was added para-toluene sulfonic acid monohydrate (12.4 g) and water (50 mL), and the reaction was heated to 65 °C for 6 days. The reaction mixture was cooled to room temperature and was quenched by the addition of saturated aqueous sodium bicarbonate and ethyl acetate. The layers were separated, and the organic layer was washed with brine. The combined aqueous layers were back-extracted with ethyl acetate. The combined organic layers were dried with sodium sulfate, filtered and concentrated under reduced pressure. The waxy solids were triturated with heptane (150 mL) to provide the title compound. MS (DCI) m/e 311.1 (M+H)+.
2.66.8 (3S,5S)-5-(((tert-butyldimethylsilyl)oxy)methyl)-3- (dibenzylamino)pyrrolidin-2-one
To a solution of Example 2.66.7 (9.3 g) and 1H-imidazole (2.2 g) in N,N-dimethylformamide was added tert-butylchlorodimethylsilane (11.2 mL, 50 weight % in toluene), and the reaction was stirred overnight. The reaction mixture was quenched by the addition of water and diethyl ether. The layers were separated, and the organic layer was washed with brine. The combined aqueous layers were back-extracted with diethyl ether. The combined organic layers were dried with sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with 35% ethyl acetate in heptane, to provide the title compound. MS (DCI) m/e 425.1 (M+H)+.
2.66.9 tert-butyl 2-((3S,5S)-5-(((tert-butyldimethylsilyl)oxy)methyl)-3- (dibenzylamino)-2-oxopyrrolidin-1-yl)acetate
To a cold (0 °C) solution of Example 2.66.8 (4.5 g) in tetrahydrofuran (45 mL) was added 95% sodium hydride (320 mg) in two portions. The cold solution was stirred for 40 minutes, and tert- butyl 2-bromoacetate (3.2 mL) was added. The reaction mixture was warmed to room temperature and stirred overnight. The reaction mixture was quenched by the addition of water and ethyl acetate. The layers were separated, and the organic layer was washed with brine. The combined aqueous layers were back-extracted with ethyl acetate. The combined organic layers were dried with sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with a gradient of 5-12% ethyl acetate in heptane, to provide the title compound. MS (DCI) m/e 539.2 (M+H)+.
2.66.10 tert-butyl 2-((3S,5S)-3-(dibenzylamino)-5-(hydroxymethyl)-2- oxopyrrolidin-1-yl)acetate
To a solution of Example 2.66.9 (5.3 g) in tetrahydrofuran (25 mL) was added
tetrabutylammonium fluoride (11 mL, 1.0M in 95/5 tetrahydrofuran /water). The reaction mixture was stirred at room temperature for one hour and was quenched by the addition of saturated aqueous ammonium chloride solution, water and ethyl acetate. The layers were separated, and the organic layer was washed with brine. The combined aqueous layers were back-extracted with ethyl acetate. The combined organic layers were dried with sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with 35% ethyl acetate in heptane, to provide the title compound. MS (DCI) m/e 425.1 (M+H)+.
2.66.11 tert-butyl 2-((3S,5S)-5-((2-((4-((tert-butyldiphenylsilyl)oxy)- 2,2-dimethylbutoxy)sulfonyl)ethoxy)methyl)-3- (dibenzylamino)-2-oxopyrrolidin-1-yl)acetate
To a solution of Example 2.66.10 (4.7 g) in dimethyl sulfoxide (14 mL) was added a solution of 4-((tert-butyldiphenylsilyl)oxy)-2,2-dimethylbutyl ethenesulfonate (14.5 g) in dimethyl sulfoxide (14 mL). Potassium carbonate (2.6 g) and water (28 µL) were added, and the reaction was heated at 60 °C under nitrogen for one day. The reaction was cooled to room temperature, and quenched by the addition of brine solution, water and diethyl ether. The layers were separated, and the organic layer was washed with brine. The combined aqueous layers were back-extracted with diethyl ether. The combined organic layers were dried with sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluting with a gradient of 15-25% ethyl acetate in heptane, to provide the title compound. MS (ESI+) m/e 871.2 (M+H)+.
2.66.12 tert-butyl 2-((3S,5S)-3-amino-5-((2-((4-((tert- butyldiphenylsilyl)oxy)-22- dimethylbutoxy)sulfonyl)ethoxy)methyl)-2-oxopyrrolidin-1- yl)acetate
Example 2.66.11 (873 mg) was dissolved in ethyl acetate (5 mL) and methanol (15 mL), and palladium hydroxide on carbon, 20% by wt (180 mg) was added. The reaction mixture was stirred under a hydrogen atmosphere (30 psi) at room temperature for 30 hours, then at 50 °C for one hour. The reaction was cooled to room temperature, filtered, and concentrated to give the desired product. MS (ESI+) m/e 691.0 (M+H)+.
2.66.13 4-(((3S,5S)-1-(2-(tert-butoxy)-2-oxoethyl)-5-((2-((4-((tert- butyldiphenylsilyl)oxy)-2,2- dimethylbutoxy)sulfonyl)ethoxy)methyl)-2-oxopyrrolidin-3- yl)amino)-4-oxobut-2-enoic acid
Maleic anhydride (100 mg) was dissolved in dichloromethane (0.90 mL), and a solution of Example 2.66.12 (650 mg) in dichloromethane (0.90 mL) was added dropwise, and then heated at 40 °C for 2 hours. The reaction mixture was directly purified by silica gel chromatography, eluting with a gradient of 1.0-2.5% methanol in dichloromethane containing 0.2% acetic acid. After concentrating the product-bearing fractions, toluene (10 mL) was added and the mixture was concentrated again to provide the title compound. MS (ESI-) m/e 787.3 (M-H)-.
2.66.14 tert-butyl 2-((3S,5S)-5-((2-((4-((tert-butyldiphenylsilyl)oxy)- 2,2-dimethylbutoxy)sulfonyl)ethoxy)methyl)-3-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)-2-oxopyrrolidin-1-yl)acetate
Example 2.66.13 (560 mg) was slurried in toluene (7 mL), and triethylamine (220 µL) and sodium sulfate (525 mg) were added. The reaction mixture was heated at reflux under a nitrogen atmosphere for 6 hours, and the reaction mixture stirred at room temperature overnight. The reaction was filtered, and the solids rinsed with ethyl acetate. The eluent was concentrated under reduced pressure, and the residue was purified by silica gel chromatography, eluting with 45/55 heptane/ethyl acetate, ethyl acetate, and then 97.5/2.5/0.2 dichloromethane/methanol/acetic acid to provide the title compound.
2.66.15 2-((3S,5S)-3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)-2-oxo-5- ((2-sulfoethoxy)methyl)pyrrolidin-1-yl)acetic acid
Example 2.66.14 (1.2 g) was dissolved in trifluoroacetic acid (15 mL) and heated to 65-70 °C under nitrogen overnight. The trifluoroacetic acid was removed under reduced pressure. The residue was dissolved in acetonitrile (2.5 mL) and purified by preparative reverse-phase liquid
chromatography on a Luna C18(2) AXIA column (250 x 50 mm, 10µ particle size) using a gradient of 5-75% acetonitrile containing 0.1% trifluoroacetic acid in water over 30 minutes, to provide the title compound. MS (ESI-) m/e 375.2 (M-H)-.
2.66.16 3-(1-((3-(2-((((4-((S)-2-((S)-2-amino-3- methylbutanamido)propanamido)-2-(2-((2S 3R 4R 5S 6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)ethyl)benzyl)oxy)carbonyl)(2-methoxyethyl)amino)ethoxy)- 5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4- yl)-6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-5-methoxy-3,4- dihydroisoquinolin-2(1H)-yl)picolinic acid
Example 1.12.10 (75 mg) and Example 2.65.19 (100 mg) were dissolved in N,N- dimethylformamide (0.3 mL). 1-Hydroxybenzotriazole (13 mg) and N-ethyl-N-isopropylpropan-2- amine (50 µL) were added, and the reaction was stirred at room temperature for two hours. The reaction mixture was concentrated under reduced pressure. The residue was dissolved in
tetrahydrofuran and methanol (0.3 mL each), and lithium hydroxide hydrate (55 mg) in water (0.6 mL) was added. The reaction mixture was stirred at room temperature for one hour and quenched by the addition of N,N-dimethylformamide/water 1/1 (1.5 mL) with trifluoroacetic acid (0.15 mL). The solution was washed with heptane (1 mL), then purified by reverse-phase chromatography (C18 column), eluting with 20-70% acetonitrile in 0.1% trifluoroacetic acid water, to provide the title compound as a trifluoroacetic acid salt. MS (ESI-) m/e 1355.6 (M-H)-.
2.66.17 (6S)-2,6-anhydro-6-[2-(2-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-2- carboxypyridin-3-yl}-5-methyl-1H-pyrazol-1-yl)methyl]-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl}oxy)ethyl](2- methoxyethyl)carbamoyl}oxy)methyl]-5-{[N-({(3S,5S)-3-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)-2-oxo-5-[(2- sulfoethoxy)methyl]pyrrolidin-1-yl}acetyl)-L-valyl-L- alanyl]amino}phenyl)ethyl]-L-gulonic acid
To a solution of Example 2.66.15 (20 mg) in N,N-dimethylformamide (0.2 mL) was added O- (7-azabenzotriazol-1-yl)-N,N,N’,N’-tetramethyluronium hexafluorophosphate (20 mg) and N,N- diisopropylethylamine (18 µL). The reaction mixture was stirred for 3 minutes at room temperature and was then added to a solution of Example 2.66.16 (57 mg) and N,N-diisopropylethylamine (30 µL) in N,N-dimethylformamide (0.7 mL). The reaction mixture was stirred at room temperature for 1 hour and diluted with N,N-dimethylformamide/water 1/1 (1.0 mL). The solution was purified by reverse-phase chromatography (C18 column), eluting with 20-70% acetonitrile in 0.1% trifluoroacetic acid water, to provide the title compound. 1H NMR (400 MHz, dimethyl sulfoxide-d6) δ ppm 9.84 (br d, 1H), 8.18 (br d, 1H), 8.04 (m, 1H), 8.01 (d, 1H), 7.77 (dd, 2H), 7.50 (d, 1H), 7.46 (m, 3H), 7.34 (t, 1H), 7.29 (s, 1H), 7.21 (br d, 1H), 7.07 (s, 2H), 7.01 (d, 1H), 6.99 (d, 1H), 5.00 (s, 4H), 4.64 (t, 1H), 4.37 (m, 1H), 4.18 (m, 2H), 4.01 (d, 1H), 3.88 (s, 3H), 3.87 (m, 2H), 3.81 (br d, 2H), 3.73 (br m, 1H), 3.63 (m, 2H), 3.55 (m, 2H), 3.49 (m, 2H), 3.36 (br m, 6H), 3.31 (m, 2H), 3.26 (br m, 2H), 3.19 (m, 2H), 3.14 (m, 1H), 3.10 (br m, 1H), 2.94 (t, 1H), 2.81 (m, 3H), 2.74 (m, 2H), 2.60 (br m, 1H), 2.36 (m, 1H), 2.09 (s, 3H), 2.00 (m, 2H), 1.85 (m, 1H), 1.55 (br m, 1H), 1.40-0.92 (m, 14H), 0.88, 0.86, 0.83, 0.79 (d,d, s, s, total 12H). MS (ESI-) m/e 1713.7 (M-1).
2.67 Synthesis of 8-[2-({[(3-amino-3-oxopropyl){2-[(3-{[4-(6-{8-[(1,3- benzothiazol-2-yl)carbamoyl]-3,4-dihydroisoquinolin-2(1H)-yl}-2- carboxypyridin-3-yl)-5-methyl-1H-pyrazol-1-yl]methyl}-5,7- dimethyltricyclo[3.3.1.13,7]decan-1-yl)oxy]ethyl}carbamoyl]oxy}methyl)- 5-{[(2S)-2-({(2S)-2-[2-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetamido]- 3-methylbutanoyl}amino)propanoyl]amino}phenyl]-2,6-anhydro-7,8- dideoxy-L-glycero-L-gulo-octonic acid (Synthon ZT)
2.67.1 3-(1-((3-(2-((((4-((S)-2-((S)-2-amino-3- methylbutanamido)propanamido)-2-(2-((3R,4R,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)ethyl)benzyl)oxy)carbonyl)(3-amino-3- oxopropyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)picolinic acid
To a cold (0 °C) solution of Example 2.65.19 (66 mg) and Example 1.32.2 (60 mg) in N,N- dimethylformamide (6 mL) was added N,N-diisopropylethylamine (0.026 mL) and
1-hydroxybenzotriazole hydrate (16.23 mg). The reaction mixture was slowly warmed to room temperature and stirred overnight. To the reaction mixture was added water (1 mL) and LiOH H2O(20 mg). The mixture was stirred at room temperature for 3 hours. The mixture was acidified with trifluoroacetic acid, filtered and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1% trifluoroacetic acid, to provide the title compound. MS (ESI) m/e 1338.5 (M-H)-.
2.67.2 8-[2-({[(3-amino-3-oxopropyl){2-[(3-{[4-(6-{8-[(1,3- benzothiazol-2-yl)carbamoyl]-3,4-dihydroisoquinolin-2(1H)- yl}-2-carboxypyridin-3-yl)-5-methyl-1H-pyrazol-1-yl]methyl}- 5,7-dimethyltricyclo[3.3.1.13,7]decan-1- yl)oxy]ethyl}carbamoyl]oxy}methyl)-5-{[(2S)-2-({(2S)-2-[2-(2,5- dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetamido]-3- methylbutanoyl}amino)propanoyl]amino}phenyl]-2,6-anhydro- 7,8-dideoxy-L-glycero-L-gulo-octonic acid
The title compound was prepared as described in Example 2.58.7, substituting Example 2.58.6 with Example 2.67.1. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ ppm 9.91 (d, 1H), 8.25 (dd, 2H), 8.03 (d, 1H), 7.79 (d, 1H), 7.61 (d, 6H), 7.55– 7.30 (m, 7H), 7.28 (s, 1H), 7.22 (d, 1H), 7.07 (s, 2H), 6.94 (d, 1H), 6.89– 6.74 (m, 1H), 5.01 (s, 3H), 4.96 (s, 2H), 4.38 (t, 1H), 4.27– 4.17 (m, 1H), 4.12 (d, 2H), 3.88 (t, 2H), 3.79 (d, 1H), 3.41– 3.30 (m, 3H), 3.24 (s, 2H), 3.12 (dt, 2H), 3.01 (t, 2H), 2.94 (t, 1H), 2.74 (d, 1H), 2.67– 2.56 (m, 1H), 2.29 (t, 2H), 2.08 (d, 3H), 1.99 (d, 3H), 1.55 (d, 1H), 1.42– 0.99 (m, 15H), 0.99– 0.70 (m, 12H). MS (ESI) m/e 1477.2 (M+H)+.
2.68 Synthesis of 4-{[({2-[(3-{[4-(6-{8-[(1,3-benzothiazol-2-yl)carbamoyl]-3,4- dihydroisoquinolin-2(1H)-yl}-2-carboxypyridin-3-yl)-5-methyl-1H- pyrazol-1-yl]methyl}-5,7-dimethyltricyclo[3.3.1.13,7]decan-1- yl)oxy]ethyl}[3-(methylamino)-3-oxopropyl]carbamoyl)oxy]methyl}-3-{3- [2-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetamido]propoxy}phenyl beta-D-glucopyranosiduronic acid (Synthon AAN)
2.68.1 3-(1-((3-(2-((((2-(3-aminopropoxy)-4-(((2S,3R,4S,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)oxy)benzyl)oxy)carbonyl)(3-(methylamino)-3- oxopropyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)picolinic acid
To a cold (0 °C) solution of Example 2.28.3 (38.7 mg) and Example 1.39 (39.3 mg) in N,N- dimethylformamide (6 mL) was added N,N-diisopropylethylamine (0.026 mL) and
1-hydroxybenzotriazole hydrate (6.58 mg). The reaction was slowly warmed to room temperature and stirred overnight. To the reaction was added water (2 mL) and LiOH H2O (50 mg), and the mixture was stirred at room temperature for 3 hours. The mixture was acidified with trifluoroacetic acid, filtered and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20- 80% acetonitrile in water containing 0.1% trifluoroacetic acid, to provide the title compound. MS (ESI) m/e 1230.2 (M-H)-.
2.68.2 4-{[({2-[(3-{[4-(6-{8-[(1,3-benzothiazol-2-yl)carbamoyl]-3,4- dihydroisoquinolin-2(1H)-yl}-2-carboxypyridin-3-yl)-5-methyl- 1H-pyrazol-1-yl]methyl}-5,7-dimethyltricyclo[3.3.1.13,7]decan- 1-yl)oxy]ethyl}[3-(methylamino)-3- oxopropyl]carbamoyl)oxy]methyl}-3-{3-[2-(2,5-dioxo-2,5- dihydro-1H-pyrrol-1-yl)acetamido]propoxy}phenyl beta-D- glucopyranosiduronic acid
The title compound was prepared as described in Example 2.58.7, substituting Example 2.58.6 with Example 2.68.1 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ ppm 12.88 (s, 2H), 9.93 (d, 1H), 8.36– 8.22 (m, 2H), 8.04 (d, 1H), 7.80 (d, 2H), 7.76 (d, 0H), 7.62 (d, 1H), 7.56– 7.42 (m, 5H), 7.41– 7.33 (m, 3H), 7.28 (s, 1H), 7.22 (d, 1H), 7.08 (s, 2H), 6.95 (d, 1H), 5.01 (d, 3H), 4.96 (s, 2H), 4.39 (p, 1H), 4.22 (dd, 1H), 4.12 (d, 2H), 3.89 (t, 2H), 3.80 (d, 2H), 3.34 (t, 2H), 3.22 (d, 2H), 3.13 (dt, 2H), 3.02 (t, 2H), 2.94 (t, 1H), 2.86– 2.71 (m, 1H), 2.60 (s, 2H), 2.54 (d, 4H), 2.29 (q, 2H), 2.09 (d, 3H), 2.07– 1.90 (m, 3H), 1.60– 1.48 (m, 1H), 1.39– 1.00 (m, 17H), 0.97– 0.74 (m, 15H). (ESI) m/e 1489.5 (M-H)-.
2.69 Synthesis of 2,6-anhydro-8-(2-{[({2-[(3-{[4-(6-{8-[(1,3-benzothiazol-2- yl)carbamoyl]-3,4-dihydroisoquinolin-2(1H)-yl}-2-carboxypyridin-3-yl)- 5-methyl-1H-pyrazol-1-yl]methyl}-5,7-dimethyltricyclo[3.3.1.13,7]decan- 1-yl)oxy]ethyl}[3-(methylamino)-3-oxopropyl]carbamoyl)oxy]methyl}-5- {[(2S)-2-({(2S)-2-[2-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetamido]-3- methylbutanoyl}amino)propanoyl]amino}phenyl)-7,8-dideoxy-L-glycero- L-gulo-octonic acid (Synthon AAO)
2.69.1 3-(1-((3-(2-((((4-((S)-2-((S)-2-amino-3- methylbutanamido)propanamido)-2-(2-((2S,3R,4R,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)ethyl)benzyl)oxy)carbonyl)(3-(methylamino)-3- oxopropyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)picolinic acid
The title compound was prepared as described in Example 2.67.1, substituting Example 1.32.2 with Example 1.39. MS (ESI) m/e 1352.6 (M-H)-.
2.69.2 2,6-anhydro-8-(2-{[({2-[(3-{[4-(6-{8-[(1,3-benzothiazol-2- yl)carbamoyl]-3,4-dihydroisoquinolin-2(1H)-yl}-2- carboxypyridin-3-yl)-5-methyl-1H-pyrazol-1-yl]methyl}-5,7- dimethyltricyclo[3.3.1.13,7]decan-1-yl)oxy]ethyl}[3- (methylamino)-3-oxopropyl]carbamoyl)oxy]methyl}-5-{[(2S)-2- ({(2S)-2-[2-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)acetamido]- 3-methylbutanoyl}amino)propanoyl]amino}phenyl)-7,8- dideoxy-L-glycero-L-gulo-octonic acid
The title compound was prepared as described in Example 2.58.7, substituting Example 2.58.6 with Example 2.67.1. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ ppm 12.88 (s, 2H), 9.93 (d, 1H), 8.36– 8.22 (m, 2H), 8.04 (d, 1H), 7.80 (d, 2H), 7.76 (d, 0H), 7.62 (d, 1H), 7.56– 7.42 (m, 5H), 7.41– 7.33 (m, 3H), 7.28 (s, 1H), 7.22 (d, 1H), 7.08 (s, 2H), 6.95 (d, 1H), 5.01 (d, 3H), 4.96 (s, 2H), 4.39 (p, 1H), 4.22 (dd, 1H), 4.12 (d, 2H), 3.89 (t, 2H), 3.80 (d, 2H), 3.34 (t, 2H), 3.22 (d, 2H), 3.13 (dt, 2H), 3.02 (t, 2H), 2.94 (t, 1H), 2.86– 2.71 (m, 1H), 2.60 (s, 2H), 2.54 (d, 4H), 2.29 (q, 2H), 2.09 (d, 3H), 2.07– 1.90 (m, 3H), 1.60– 1.48 (m, 1H), 1.39– 1.00 (m, 17H), 0.97– 0.74 (m, 15H). MS (ESI) m/e 1489.5 (M-H)-.
2.70 Synthesis of 2,6-anhydro-8-(2-{[({2-[(3-{[4-(6-{8-[(1,3-benzothiazol-2- yl)carbamoyl]-3,4-dihydroisoquinolin-2(1H)-yl}-2-carboxypyridin-3-yl)- 5-methyl-1H-pyrazol-1-yl]methyl}-57-dimethyltricyclo[331137]decan- 1-yl)oxy]ethyl}[3-(methylamino)-3-oxopropyl]carbamoyl)oxy]methyl}-5- {[(2S)-2-{[(2S)-2-(2-{(3S,5S)-3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)-2- oxo-5-[(2-sulfoethoxy)methyl]pyrrolidin-1-yl}acetamido)-3- methylbutanoyl]amino}propanoyl]amino}phenyl)-7,8-dideoxy-L-glycero- L-gulo-octonic acid (Synthon AAP)
To a solution of Example 2.66.15 (17 mg) in N,N-dimethylformamide (320 µL) was added O- (7-azabenzotriazol-1-yl)-N,N,N’,N’-tetramethyluronium hexafluorophosphate (19 mg) and N,N- diisopropylethylamine (17 µL). The reaction mixture was stirred for 5 minutes and was added to a solution of Example 2.69.1 (39 mg) and N,N-diisopropylethylamine (36 µL) in N,N- dimethylformamide (320 µL). The reaction mixture was stirred for 2 hours and was diluted with N,N- dimethylformamide (2 mL). The solution was filtered and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1%
trifluoroacetic acid, to provide the title compound. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ ppm 9.82 (s, 1H), 8.15 (d, 1H), 8.00 (dd, 2H), 7.75 (d, 1H), 7.58 (d, 1H), 7.44 (ddd, 5H), 7.32 (td, 2H), 7.25 (s, 1H), 7.18 (d, 1H), 7.03 (s, 2H), 6.92 (d, 1H), 6.76 (s, 1H), 4.97 (s, 2H), 4.92 (s, 2H), 4.61 (t, 1H), 4.33 (p, 1H), 4.21– 4.08 (m, 2H), 3.98 (d, 1H), 3.84 (t, 2H), 3.40– 3.27 (m, 3H), 3.21 (s, 1H), 3.14– 3.03 (m, 2H), 2.98 (t, 2H), 2.90 (t, 1H), 2.81– 2.50 (m, 4H), 2.38– 2.20 (m, 3H), 2.05 (s, 3H), 2.01– 1.90 (m, 2H), 1.88– 1.74 (m, 1H), 1.60– 1.43 (m, 1H), 1.36– 0.95 (m, 14H), 0.95– 0.62 (m, 13H). MS (ESI) m/e 1710.5(M-H)-.
2.71 Synthesis of 6-{8-[(1,3-benzothiazol-2-yl)carbamoyl]-3,4- dihydroisoquinolin-2(1H)-yl}-3-[1-({3-[2-({[(4-{[(2S)-5- (carbamoylamino)-2-{[(2S)-2-{[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanoyl]amino}-3- methylbutanoyl]amino}pentanoyl]amino}phenyl)methoxy]carbonyl}ami no)acetamido]-5,7-dimethyltricyclo[3.3.1.13,7]decan-1-yl}methyl)-5- methyl-1H-pyrazol-4-yl]pyridine-2-carboxylic acid (Synthon ABF) The title compound was prepared as described in Example 2.2, substituting Example 1.3.2 with Example 1.40.11. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ ppm 9.96 (s, 1H), 8.03 (dd, 2H), 7.78 (d, 2H), 7.59 (dd, 3H), 7.53– 7.39 (m, 3H), 7.35 (q, 2H), 7.30– 7.23 (m, 3H), 7.20 (d, 1H), 6.98 (s, 2H), 6.94 (d, 1H), 4.94 (d, 4H), 4.38 (t, 1H), 4.17 (dd, 1H), 3.87 (t, 2H), 3.78 (s, 2H), 3.35 (t, 2H), 3.00 (t, 3H), 2.94 (s, 0H), 2.16 (d, 1H), 2.09 (s, 3H), 1.95 (d, 1H), 1.74– 1.27 (m, 10H), 1.13 (dq, 5H), 0.87– 0.71 (m, 12H). MS (ESI) m/e 1355.5(M-H)-.
2.72 Synthesis of 8-[2-({[(3-amino-3-oxopropyl){2-[(3-{[4-(6-{8-[(1,3- benzothiazol-2-yl)carbamoyl]-3,4-dihydroisoquinolin-2(1H)-yl}-2- carboxypyridin-3-yl)-5-methyl-1H-pyrazol-1-yl]methyl}-5,7- dimethyltricyclo[3.3.1.13,7]decan-1-yl)oxy]ethyl}carbamoyl]oxy}methyl)- 5-{[(2S)-2-{[(2S)-2-(2-{(3S 5S)-3-(25-dioxo-25-dihydro-1H-pyrrol-1-yl)- 2-oxo-5-[(2-sulfoethoxy)methyl]pyrrolidin-1-yl}acetamido)-3- methylbutanoyl]amino}propanoyl]amino}phenyl]-2,6-anhydro-7,8- dideoxy-L-glycero-L-gulo-octonic acid (Synthon ZZ)
2.72.1 3-(1-((3-(2-((((4-((S)-2-((S)-2-amino-3- methylbutanamido)propanamido)-2-(2-((3R,4R,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)ethyl)benzyl)oxy)carbonyl)(3-amino-3- oxopropyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6-(8-(benzo[d]thiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)picolinic acid
To a cold (0 °C) solution of Example 2.65.19 (66 mg) and Example 1.32.2 (6 mL) were added N,N-diisopropylamine (0.026 mL) and 1-hydroxybenzotriazole hydrate (16.23 mg). The reaction mixture was slowly warmed to room temperature and stirred overnight. To the reaction mixture was added water (1 mL) and LiOH H2O (20 mg), and the mixture was stirred at room temperature for 3 hours. The mixture was acidified with trifluoroacetic acid, filtered and was purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1% trifluoroacetic acid, to provide the title compound. MS (ESI) m/e 1338.5 (M-H)-.
2.72.2 8-[2-({[(3-amino-3-oxopropyl){2-[(3-{[4-(6-{8-[(1,3- benzothiazol-2-yl)carbamoyl]-3,4-dihydroisoquinolin-2(1H)- yl}-2-carboxypyridin-3-yl)-5-methyl-1H-pyrazol-1-yl]methyl}- 5,7-dimethyltricyclo[3.3.1.13,7]decan-1- yl)oxy]ethyl}carbamoyl]oxy}methyl)-5-{[(2S)-2-{[(2S)-2-(2- {(3S,5S)-3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)-2-oxo-5-[(2- sulfoethoxy)methyl]pyrrolidin-1-yl}acetamido)-3- methylbutanoyl]amino}propanoyl]amino}phenyl]-2,6-anhydro- 7,8-dideoxy-L-glycero-L-gulo-octonic acid
To a solution of 2-((3S,5S)-3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)-2-oxo-5-((2- sulfoethoxy)methyl)pyrrolidin-1-yl)acetic acid (17 mg) in N,N-dimethylformamide (320 µL), was added O-(7-azabenzotriazol-1-yl)-N,N,N’,N’-tetramethyluronium hexafluorophosphate (19 mg) and N-ethyl-N-isopropylpropan-2-amine (17 µL). The reaction mixture was stirred for 5 minutes and was added to a solution of Example 2.72.1 (50 mg) and N-ethyl-N-isopropylpropan-2-amine (36 µL) in N,N-dimethylformamide (320 µL). The reaction mixture was stirred for 2 hours. The reaction mixture was diluted with N,N-dimethylformamide/water (1/1, 1 mL) and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1% trifluoroacetic acid, to provide the title compound. 1H NMR (501 MHz, dimethyl sulfoxide-d6) δ ppm 9.82 (s, 1H), 8.15 (d, 1H), 8.00 (dd, 2H), 7.75 (d, 1H), 7.58 (d, 1H), 7.44 (ddd, 5H), 7.32 (td, 2H), 725 (s 1H) 718 (d 1H) 703 (s 2H) 692 (d 1H) 676 (s 1H) 497 (s 2H) 492 (s 2H) 461 (t 1H), 4.33 (p, 1H), 4.21– 4.08 (m, 2H), 3.98 (d, 1H), 3.84 (t, 2H), 3.40– 3.27 (m, 3H), 3.21 (s, 1H), 3.14– 3.03 (m, 2H), 2.98 (t, 2H), 2.90 (t, 1H), 2.81– 2.50 (m, 4H), 2.38– 2.20 (m, 3H), 2.05 (s, 3H), 2.01– 1.90 (m, 2H), 1.88– 1.74 (m, 1H), 1.60– 1.43 (m, 1H), 1.36– 0.95 (m, 14H), 0.95– 0.62 (m, 13H). MS (ESI) m/e 1697.5 (M-H)-.
2.73 Synthesis of N-[6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)hexanoyl]-L- valyl-N-{4-[({[2-({3-[(4-{6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]-2-carboxypyridin-3-yl}-5-methyl-1H- pyrazol-1-yl)methyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl}oxy)ethyl](2-sulfoethyl)carbamoyl}oxy)methyl]phenyl}-N5- carbamoyl-L-ornithinamide (Synthon CZ)
Example 1.44.2 (100 mg) and 4-((S)-2-((S)-2-(6-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)hexanamido)-3-methylbutanamido)-5-ureidopentanamido)benzyl (4-nitrophenyl) carbonate (purchased from Synchem, 114 mg) in N,N-dimethylformamide (7 mL) was cooled in an water-ice bath, and N,N-diisopropylethylamine (0.15 mL) was added. The mixture was stirred at 0 oC for 30 minutes and then at room temperature overnight. The reaction was purified by a reverse phase HPLC using a Gilson system, eluting with 20-60% acetonitrile in water containing 0.1% v/v trifluoroacetic acid, to provide the title compound. 1H NMR (400 MHz, dimethylsulfoxide-d6) δ ppm 12.85 (s, 1H), 9.99 (s, 1H), 8.04 (t, 2H), 7.75-7.82 (m, 2H), 7.40-7.63 (m, 6H), 7.32-7.39 (m, 2H), 7.24-7.29 (m, 3H), 6.99 (s, 2H), 6.95 (d, 1H), 6.01 (s, 1H), 4.83-5.08 (m, 4H), 4.29-4.48 (m, 1H), 4.19 (t, 1H), 3.84- 3.94 (m, 2H), 3.80 (d, 2H), 3.14-3.29 (m, 2H), 2.87-3.06 (m, 4H), 2.57-2.69 (m, 2H), 2.03-2.24 (m, 5H), 1.89-2.02 (m, 1H), 1.53-1.78 (m, 2H), 1.26-1.53 (m, 8H), 0.89-1.27 (m, 12H), 0.75-0.88 (m, 12H). MS (ESI) m/e 1452.2 (M+H)+.
2.74 Synthesis of 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)- yl)-3-(1-((3-(2-((((2-(2-((2S,3R,4R,5S,6S)-6-carboxy-3,4,5-trihydroxytetrahydro- 2H-pyran-2-yl)ethyl)-4-((S)-2-((S)-2-(2-((3S,5S)-3-(2,5-dioxo-2,5-dihydro-1H- pyrrol-1-yl)-2-oxo-5-((2-sulfoethoxy)methyl)pyrrolidin-1-yl)acetamido)-3- methylbutanamido)propanamido)benzyl)oxy)carbonyl)(2- sulfoethyl)amino)ethoxy)-5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H- pyrazol-4-yl)picolinic acid (Synthon TX)
2.74.1 3-(1-(((1r,3s,5R,7S)-3-(2-((((4-((R)-2-((R)-2-amino-3- methylbutanamido)propanamido)-2-(2-((2S,3R,4R,5S,6S)-6- carboxy-3,4,5-trihydroxytetrahydro-2H-pyran-2- yl)ethyl)benzyl)oxy)carbonyl)(2-sulfoethyl)amino)ethoxy)-5,7- dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol-4-yl)-6- (8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)picolinic acid To a cold (0 °C) solution of Example 2.65.19 (70 mg) and Example 1.44.2 (58.1 mg) in N,N- dimethylformamide (4 mL) was added N-ethyl-N-isopropylpropan-2-amine (0.026 mL). The reaction was slowly warmed to room temperature and stirred overnight. To the reaction mixture was added water (1 mL) and LiOH H2O (20 mg). The mixture was stirred at room temperature for 3 hours. The mixture was acidified with trifluoroacetic acid, filtered and purified by reverse-phase HPLC on a Gilson system (C18 column), eluting with 20-80% acetonitrile in water containing 0.1%
trifluoroacetic acid, to give the title product.
MS (ESI) m/e 1564.4 (M-H)-.
2.74.2 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl)-3-(1-((3-(2-((((2-(2-((2S,3R,4R,5S,6S)-6-carboxy- 3,4,5-trihydroxytetrahydro-2H-pyran-2-yl)ethyl)-4-((S)-2-((S)- 2-(2-((3S,5S)-3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)-2-oxo-5- ((2-sulfoethoxy)methyl)pyrrolidin-1-yl)acetamido)-3- methylbutanamido)propanamido)benzyl)oxy)carbonyl)(2- sulfoethyl)amino)ethoxy)-5,7-dimethyladamantan-1- yl)methyl)-5-methyl-1H-pyrazol-4-yl)picolinic acid
The title compound was prepared by substituting Example 2.74.1 for Example 2.66.16 in Example 2.66.17.1H NMR (400 MHz, dimethylsulfoxide-d6) δ ppm 9.85 (s, 1H), 8.17 (br d, 1H), 8.01 (d, 2H), 7.77 (d, 1H), 7.59 (d, 1H), 7.53 (d, 1H), 7.43 (m, 4H), 7.34 (m, 3H), 7.19 (d, 1H), 7.06 (s, 2H), 6.96 (d, 1H), 4.99 (m, 2H), 4.95 (s, 2H), 4.63 (t, 1H), 4.36 (t, 1H), 4.19 (br m, 1H), 4.16 (d, 1H), 3.98 (d, 1H), 3.87 (br t, 2H), 3.81 (br d, 2H), 3.73 (brm, 1H), 3.63 (t, 2H), 3.53 (m, 2H), 3.44 (m, 4H), 3.31 (t, 2H), 3.21 (br m, 2H), 3.17 (m, 2H), 3.00 (m, 2H), 2.92 (br m, 1H), 2.75 (m, 3H), 2.65 (br m, 3H), 2.35 (br m, 1H), 2.07 (s, 3H), 1.98 (br m, 2H), 1.85 (m, 1H), 1.55 (br m, 1H), 1.34 (br m, 1H), 1.26 (br m, 6H), 1.09 (br m, 7H), 0.93 (br m, 1H), 0.87, 0.83, 0.79 (all d, total 12H). MS (ESI) m/e 1733.4 (M-H)-. Example 3: Generation of Mouse Anti-B7-H3 Monoclonal Antibodies by Mouse Hybridoma Technology
B7-H3 specific antibodies were raised using mouse hybridomas technology. Specifically, a mouse fibroblast cell line (3T12) expressing full length human B7-H3 as well as recombinant human or mouse B7-H3-ECD-human Fc fusion proteins were used as immunogens, the sequences of which are provided in Table 1. Human HCT116 cell lines expressing human B7-H3 were used for determining anti-sera titer and for screening antigen-specific antibodies. Cell lines were exposed to approximately 3000 mREM of gamma source radiation prior to immunization. Two different strains of mice were immunized in the hock with dosages containing 5 x 106 cells/mouse/injection or 10 ug of protein/mouse/injection in the presence of Gerbu MM adjuvant (Cooper-Casey Corporation, Valley Center, CA, US) for both primary and boost immunizations. To increase immune response to mouse B7-H3, the mice were further boosted with a mixture of human and mouse B7-H3-ECD-human Fc proteins for the final boosts. Briefly, the antigens were prepared in PBS as follows: 200 x 106 cells/mL or 400 ug/mL protein. The calculated volume of antigen was transferred to a sterile microcentrifuge tube and equal volume of Gerbu MM was then added. The solution was mixed by gently vortexing for 1 minute. The adjuvant-antigen solution was then drawn into a proper syringe for animal injection. A total of 25 µL of the mixture was injected into the hock of each leg of the mouse. Each animal was boosted 3 times before serum titer was determined for the groups. All animals were given 2 additional boosts with an equal mixture of mouse B7-H3-ECD-human Fc and human B7-H3- ECD-human Fc proteins in adjuvant before fusion.
Table 1: Amino acid sequences of recombinant proteins used for immunization or screening
Figure imgf000470_0001
Figure imgf000471_0001
Hybridoma fusion and screening
Cells of murine myeloma cell line (NS-0, ECACC No.85110503) were cultured to reach the log phase stage right before fusion. Popliteal and inguinal lymph nodes were removed from each mouse and single cell suspensions were prepared sterilely. Lymphocytes were fused with myeloma cells (E. Harlow, D. Lane, Antibody: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1998); Kohler G. and Milstein C.,“Continuous cultures of fused cell secreting antibody of predefined specificity,” Nature, 256:495-497 (1975); BTX Harvard Apparatus (Holliston, MA, US) ECM 2001 technical manual). Fused hybrid cells were dispensed into 96-well plates in DMEM/10% FBS/HAT media. Supernatants from surviving hybridoma colonies were subjected to cell-based screening using human cell lines expressing the recombinant human B7-H3. Briefly, a human cell line expressing the human B7-H3 was thawed and directly dispensed into 96 well (black with clear bottom for imaging) plates at 50,000 cells/well in growth media and incubated for 2 days at 37 °C to reach 50% confluency. Hybridoma supernatants (50 µL/well) were transferred to respective plates and incubated at room temperature for 30 minutes. Media was removed from each well and goat anti-mouse IgG-AF488 (Invitrogen, No. A11029, Grand Island, NY, US) was used for detection using the InCell Analyzer 2000 (GE). Hits were expanded and binding was confirmed by FACS using a different human cell line or a mouse cell line expressing the human B7-H3 and goat anti-mouse IgG-PE for detection. Species specificity was determined using the ELISA format according to the following procedure. ELISA plates were coated with human B7-H3-ECD-human Fc, cynomolgous B7-H3-ECD-his, or mouse B7-H3-ECD-human Fc proteins overnight at room temperature. Plates were washed and hybridoma supes (100 µL) was added to each well, and incubated at room temperature for 1 hour. Plates were washed, donkey anti-mouse IgG-HRP (Jackson Immunochemicals, No.115-035-071, West Grove, PA, US) was used for detection, and binding ODs were observed at 650 nm.
A selection of hits were subcloned using the MoFlo (Beckman, Indianapolis, IN, US) by depositing a single cell per well into 96 well cell culture plates to ensure clonality of the cell line. Resulting colonies were screened for specificity by FACS using mouse 3T12 fibroblast cell lines expressing the human B7-H3, cynomolgous B7-H3 or mouse B7-H3. Isotype of each monoclonal antibody was determined using the Mouse Monoclonal Isostyping Kit (Roche, No.11-493-027-001, Indianapolis, IN, USA). Hybridoma clones producing antibodies that showed high specific binding activity against human and cynomolgus B7-H3 antigen were subcloned and purified (Table 2). Table 2: List of Anti-B7-H3 antibodies enerated usin mouse h bridoma technolo
Figure imgf000472_0001
Example 4: In Vitro Characterization of Anti-B7-H3 Mouse Monoclonal Antibodies.
The binding affinity of the purified anti-B7-H3 monoclonal antibodies was determined by surface plasma resonance. Table 3 shows the association rate constants (ka) dissociation rate constants (kd) and equilibrium dissociation constants (KD) for a series of mouse hybridoma derived anti-B7-H3 monoclonal antibodies (mAbs) binding to the soluble ECDs of human B7-H3 and cyno B7-H3. The binding kinetics were derived from SPR measurements using a Biacore T200 instrument and a mAb capture approach (as described in the materials and methods below). Table 3: Biacore kinetics of anti-B7-H3 mouse hybridoma antibodies binding to human and cynomolgus monkey B7-H3.
Figure imgf000473_0001
Pair-wise binding assays performed on Biacore T200 SPR instruments were used to determine the relative epitope grouping for the murine anti-B7-H3 mAbs as described in the methods below. Figure 1 shows an epitope grouping depiction, which describes the relative human B7-H3 epitope diversity and overlap for a series of anti-B7-H3 mAbs identified herein. Epitope groups are represented as individual ovals, some of which overlap with each other. Antibodies in different epitope groups can bind to B7-H3 simultaneously and likely bind to different epitopes while antibodies within a given epitope group cannot bind to B7-H3 simultaneously and likely bind to overlapping epitopes. The grouping information was derived from a simultaneous binding assay as described in materials and methods. Ab3, Ab4, Ab5, Ab11, Ab12, and Ab8 groupings were ambiguous
Materials and methods: Binding kinetics
Biacore T200 SPR instruments were used to measure the binding kinetics of human B7-H3 (4Ig- B7-H3 variant) (analyte) binding to various mAbs (ligands). The assay format was Fc-based capture via immobilized anti-mouse (Fc) (Pierce 31170) or immobilized anti-human (Fc) (Pierce 31125). A standard amine coupling protocol was employed to immobilize the capture reagents via primary amines to the carboxy-methyl (CM) dextran surface of CM5 sensorchips (Biacore); capture antibodies were coupled to a level of approximately 5000RU. For binding kinetic measurements the assay buffer was HBS-EP+ (Biacore): 10 mM Hepes, pH7.4, 150 mM NaCl, 3 mM EDTA, 0.05% polysorbate 20. During the assay, all measurements were referenced against the capture surface alone. Each assay cycle consisted of the following steps: 1) Capture of ligand to approximately 50RU; 2) Analyte injection over both reference and test surface, 240 µL at 80 µL/min, after which the dissociation was monitored for 900 seconds at 80 µL/min; 3) Regeneration of capture surface with low pH glycine. For kinetic determinations analyte injections were 3-point, 9-fold dilution series of 900 nM, 100 nM and 11.11 nM, buffer only injections were included for secondary referencing. Data were processed and fit to a 1:1 binding model using Biacore T200 Evaluation Software to determine the binding kinetic rate constants, ka (on-rate) and kd (off-rate), and the equilibrium dissociation constant (affinity, KD). Materials and methods: Epitope grouping
Pair-wise binding assays performed on Biacore T200 SPR instruments were used to determine the relative epitope grouping for a series of anti-B7-H3 mAbs. The assay format was Fc- based capture via immobilized anti-mouse(Fc) (Pierce 31170) or immobilized anti-human (Fc) (Pierce 31125). A standard amine coupling protocol was employed to immobilize the capture reagents via primary amines to the carboxy-methyl (CM) dextran surface of CM5 sensorchips (Biacore); capture antibodies were coupled to a level of approximately 2000RU. Epitope grouping measurements were done at 12 °C (low temperature allows for grouping information on fast off-rate mAbs), the assay buffer was HBS-EP+ (Biacore): 10 mM Hepes, pH7.4, 150 mM NaCl, 3 mM EDTA, 0.05% polysorbate 20. Each assay cycle consisted of the following steps in a four flowcell system: 1) separate test mAbs were captured in flowcells 2, 3 & 4 (flowcell 1 was reference, no test mAb); 2) all 4 flowcells were then blocked by injection with isotype control mAb or isotype mAb cocktail at 50 µg/mL; 3) all 4 flowcells were then injected with antigen or buffer only (buffer only is for double referencing, done for each mAb pair individually); 4) all 4 flowcells were then injected with 2nd test mAb at 10 µg/mL; 5) all 4 flowcells were then regenerated with glycine, pH1.5. The assay was done for each test mAb pair in reciprocal orientations. Simultaneous binding was evaluated examining the ratio of the 2nd test mAb response to the Ag response (RUmAb2/RUAg); if this ratio was equal to or greater than 0.2 the interaction was scored as a simultaneous binder. From this pair-wise binding assay data a“venn” style diagram was constructed manually to depict relative epitope groupings. Example 5: Generation of Anti-hB7-H3 Chimeric Antibodies
Following the identification of mouse anti-B7-H3 hybridoma antibodies, heavy and light chain variable regions (VH and VL) corresponding to the secreted antibodies were determined from cells using reverse transcriptase-polymerase chain reaction (RT-PCR). Murine variable regions were expressed in mammalian host cells in the context of a human immunoglobulin constant region to provide chimeric antibodies. Table 4 below provides the variable region amino acid sequences for the mouse chimerized hybridomas.
Table 4: Variable region amino acid sequences of anti-B7-H3 antibodies from mouse hybridomas
Figure imgf000475_0001
Figure imgf000476_0001
Figure imgf000477_0001
Figure imgf000478_0001
Figure imgf000479_0001
Figure imgf000480_0001
Figure imgf000481_0001
Figure imgf000482_0001
Example 6: Binding Characterization of Chimeric Anti-B7-H3 Antibodies
To generate purified chimeric antibodies, expression vectors were transiently transfected into HEK2936E suspension cell cultures in a ratio of 60% to 40% light to heavy chain construct. 1 mg/ml polyethylenimine (PEI) or 2.6 µL/mL of Expifectamine was used to transfect the cells. Cell supernatants were harvested after five days in shaking flasks, spun down to pellet cells, and filtered through 0.22 µm filters to separate IgG from culture contaminants. Antibody-containing supernatants were purified on Akta Pure using protein A mAb SelectSure. Columns were equilibrated in PBS pH 7.4, supernatants were then passed through the column and a wash was performed with PBS pH 7.4. IgG were eluted with 0.1 M acetic acid pH 3.5 and collected in several aliquots. Fractions containing IgG were pooled and dialyzed in PBS overnight at 4 °C. Anti-B7-H3 chimeric antibodies that were successfully expressed were characterized for the ability to bind the B7-H3 overexpressing human non-small cell lung cancer cell line NCI-H1650 (ATCC® No. CRL-5883) by FACS using the methods described below. Table 5 summarizes the binding properties of the chimeric anti-B7-H3 antibodies. TABLE 5: In vitro characterization of B7-H3 chimeric antibodies
Figure imgf000483_0001
FACS binding methods
Cells were harvested from flasks when approximately 80% confluent using Gibco® Cell Dissociation Buffer. Cells were washed once in PBS/1% FBS (FACS buffer) then resuspended at 2.5x106 cells/mL in FACS buffer. 100 µL of cells/well were added to a round bottom 96-well plate. 10 µL of a 10x concentration of mAb/ADC (final concentrations are indicated the figures). Wells were washed twice with FACS buffer and resuspended in 50 µL of secondary Ab (AlexaFluor 488) diluted in FACS buffer. The plate was incubated at 4°C for one hour and washed twice with FACS buffer. Cells were resuspended in 100 µL of PBS/1% formaldehyde and analyzed on a Becton Dickinson LSRII flow cytometer. Data was analyzed using WinList flow cytometry analysis software. Example 7: Characterization of Chimeric Anti-B7-H3 Antibodies as Bcl-xL Inhibiting Antibody Drug Conjugates
Nine anti-B7-H3 chimeric antibodies were conjugated to the Bcl-xL inhibiting (Bcl-xLi) synthon CZ (Example 2.1) using conjugation Method A described below. The resulting ADCs (anti- B7-H3 antibodies conjugated to synthon CZ) were tested for binding to cell surface human B7-H3 by FACS (as described in Example 6) and for cell cytotoxicity in cell lines expressing B7-H3. Of the nine antibodies, three antibodies (chAb2, chAb6, and chAb16) precipitated following conjugation to synthon CZ and showed weak cytotoxicity in cells expressing human B7-H3. Table 6 provides cell surface binding and cytotoxicity activity of anti-B7-H3 chimera ADCs against breast cancer cell HCC38 expressing human B7-H3. TABLE 6: In vitro characterization of B7-H3 chimeric-CZ conjugates
Figure imgf000484_0001
Materials and methods: Conjugation of Bcl-xL inhibitory ADCs
ADCs were synthesized using one of the methods described below. Exemplary ADCs were synthesized using one of nine exemplary methods, described below. Method A. A solution of Bond-Breaker™ tris(2-carboxyethyl)phosphine hydrochloride (TCEP) solution (10 mM, 0.017 mL) was added to a solution of antibody (10 mg/mL, 1 mL) preheated to 37 °C. The reaction mixture was kept at 37 °C for 1 hour. The solution of reduced antibody was added to a solution of synthon (3.3 mM, 0.160 mL in DMSO) and gently mixed for 30 minutes. The reaction solution was loaded onto a desalting column (PD10, washed with Dulbecco’s phosphate- buffered saline [DPBS] 3x before use), followed by DPBS (3 mL). The purified ADC solution was filtered through a 0.2 micron, low protein-binding 13 mm syringe-filter and stored at 4 °C. Method B. A solution of Bond-Breaker™ tris(2-carboxyethyl)phosphine (TCEP) solution (10 mM, 0.017 mL) was added to the solution of antibody (10 mg/mL, 1 mL) preheated to 37 °C. The reaction mixture was kept at 37 °C for 1 hour. The solution of reduced antibody was adjusted to pH=8 by adding boric buffer (0.05 mL, 0.5 M, pH 8), added to a solution of synthon (3.3 mM, 0.160 mL in DMSO) and gently mixed for 4 hours. The reaction solution was loaded onto a desalting column (PD10, washed with DPBS 3x before use), followed by DPBS (1.6 mL) and eluted with additional DPBS (3 mL). The purified ADC solution was filtered through a 0.2 micron, low protein-binding 13 mm syringe-filter and stored at 4 °C. Method C. Conjugations were performed using a PerkinElmer Janus (part AJL8M01) robotic liquid handling system equipped with an I235/96 tip ModuLar Dispense Technology (MDT), disposable head (part 70243540) containing a gripper arm (part 7400358), and an 8-tip Varispan pipetting arm (part 7002357) on an expanded deck. The PerkinElmer Janus system was controlled using the WinPREP version 4.8.3.315 Software. A Pall Filter plate 5052 was pre-wet with 100 µL 1x DPBS using the MDT. Vacuum was applied to the filter plate for 10 seconds and was followed by a 5 second vent to remove DPBS from filter plate. A 50% slurry of Protein A resin (GE MabSelect Sure) in DPBS was poured into an 8 well reservoir equipped with a magnetic ball, and the resin was mixed by passing a traveling magnet underneath the reservoir plate. The 8 tip Varispan arm, equipped with 1 mL conductive tips, was used to aspirate the resin (250 µL) and transfer to a 96-well filter plate. A vacuum was applied for 2 cycles to remove most of the buffer. Using the MDT, 150 µL of 1xPBS was aspirated and dispensed to the 96-well filter plate holding the resin. A vacuum was applied, removing the buffer from the resin. The rinse/vacuum cycle was repeated 3 times. A 2 mL, 96-well collection plate was mounted on the Janus deck, and the MDT transferred 450 µL of 5x DPBS to the collection plate for later use. Reduced antibody (2 mg) as a solution in (200 µL) DPBS was prepared as described above for Conditions A and preloaded into a 96 well plate. The solutions of reduced antibody were transferred to the filter plate wells containing the resin, and the mixture was mixed with the MDT by repeated
aspiration/dispensation of a 100 µL volume within the well for 45 seconds per cycle. The aspiration/dispensation cycle was repeated for a total of 5 times over the course of 5 minutes. A vacuum was applied to the filter plate for 2 cycles, thereby removing excess antibody. The MDT tips were rinsed with water for 5 cycles (200 µL, 1 mL total volume). The MDT aspirated and dispensed 150 µL of DPBS to the filter plate wells containing resin–bound antibody, and a vacuum was applied for two cycles. The wash and vacuum sequence was repeated two more times. After the last vacuum cycle, 100 µL of 1x DPBS was dispensed to the wells containing the resin-bound antibody. The MDT then collected 30 µL each of 3.3 mM dimethyl sulfoxide solutions of synthons plated in a 96-well format and dispensed it to the filter plate containing resin-bound antibody in DPBS. The wells containing the conjugation mixture were mixed with the MDT by repeated aspiration/dispensation of a 100 µL volume within the well for 45 seconds per cycle. The aspiration/dispensation sequence was repeated for a total of 5 times over the course of 5 minutes. A vacuum was applied for 2 cycles to remove excess synthon to waste. The MDT tips were rinsed with water for 5 cycles (200 µL, 1 mL total volume). The MDT aspirated and dispensed DPBS (150 µL) to the conjugation mixture, and a vacuum was applied for two cycles. The wash and vacuum sequence was repeated two more times. The MDT gripper then moved the filter plate and collar to a holding station. The MDT placed the 2 mL collection plate containing 450 µL of 10x DPBS inside the vacuum manifold. The MDT reassembled the vacuum manifold by placement of the filter plate and collar. The MDT tips were rinsed with water for 5 cycles (200 µL, 1 mL total volume). The MDT aspirated and dispensed 100 µL of IgG Elution Buffer 3.75 (Pierce) to the conjugation mixture. After one minute, a vacuum was applied for 2 cycles, and the eluent was captured in the receiving plate containing 450 µL of 5x DPBS. The aspiration/dispensation sequence was repeated 3 additional times to deliver ADC samples with concentrations in the range of 1.5-2.5 mg/mL at pH 7.4 in DPBS. Method D. Conjugations were performed using a PerkinElmer Janus (part AJL8M01) robotic liquid handling system equipped with an I235/96 tip ModuLar Dispense Technology (MDT), disposable head (part 70243540) containing a gripper arm (part 7400358), and an 8-tip Varispan pipetting arm (part 7002357) on an expanded deck. The PerkinElmer Janus system was controlled using the WinPREP version 4.8.3.315 Software.
A Pall Filter plate 5052 was prewet with 100 µL 1x DPBS using the MDT. Vacuum was applied to the filter plate for 10 seconds and was followed by a 5 second vent to remove DPBS from filter plate. A 50% slurry of Protein A resin (GE MabSelect Sure) in DPBS was poured into an 8-well reservoir equipped with a magnetic ball, and the resin was mixed by passing a traveling magnet underneath the reservoir plate. The 8 tip Varispan arm, equipped with 1 mL conductive tips, was used to aspirate the resin (250 µL) and transfer to a 96-well filter plate. A vacuum was applied to the filter plate for 2 cycles to remove most of the buffer. The MDT aspirated and dispensed 150 µL of DPBS to the filter plate wells containing the resin. The wash and vacuum sequence was repeated two more times. A 2 mL, 96-well collection plate was mounted on the Janus deck, and the MDT transferred 450 µL of 5x DPBS to the collection plate for later use. Reduced antibody (2 mg) as a solution in (200 µL) DPBS was prepared as described above for Conditions A and dispensed into the 96-well plate. The MDT then collected 30 µL each of 3.3 mM dimethyl sulfoxide solutions of synthons plated in a 96-well format and dispensed it to the plate loaded with reduced antibody in DPBS. The mixture was mixed with the MDT by twice repeated aspiration/dispensation of a 100 µL volume within the well. After five minutes, the conjugation reaction mixture (230 µL) was transferred to the 96-well filter plate containing the resin. The wells containing the conjugation mixture and resin were mixed with the MDT by repeated aspiration/dispensation of a 100 µL volume within the well for 45 seconds per cycle. The aspiration/dispensation sequence was repeated for a total of 5 times over the course of 5 minutes. A vacuum was applied for 2 cycles to remove excess synthon and protein to waste. The MDT tips were rinsed with water for 5 cycles (200 µL, 1 mL total volume). The MDT aspirated and dispensed DPBS (150 µL) to the conjugation mixture, and a vacuum was applied for two cycles. The wash and vacuum sequence was repeated two more times. The MDT gripper then moved the filter plate and collar to a holding station. The MDT placed the 2 mL collection plate containing 450 µL of 10x DPBS inside the vacuum manifold. The MDT reassembled the vacuum manifold by placement of the filter plate and collar. The MDT tips were rinsed with water for 5 cycles (200 µL, 1 mL total volume). The MDT aspirated and dispensed 100 µL of IgG Elution Buffer 3.75 (P) to the conjugation mixture. After one minute, a vacuum was applied for 2 cycles, and the eluent was captured in the receiving plate containing 450 µL of 5x DPBS. The aspiration/dispensation sequence was repeated 3 additional times to deliver ADC samples with concentrations in the range of 1.5-2.5 mg/mL at pH 7.4 in DPBS. Method E. A solution of Bond-Breaker™ tris(2-carboxyethyl)phosphine (TCEP) solution (10 mM, 0.017 mL) was added to the solution of antibody (10 mg/mL, 1 mL) at room temperature. The reaction mixture was heated to 37 °C for 75 minutes. The solution of reduced antibody cooled to room temperature and was added to a solution of synthon (10 mM, 0.040 mL in DMSO) followed by addition of boric buffer (0.1 mL, 1M, pH 8). The reaction solution was let to stand for 3 days at room temperature, loaded onto a desalting column (PD10, washed with DPBS 3x5mL before use), followed by DPBS (1.6 mL) and eluted with additional DPBS (3 mL). The purified ADC solution was filtered through a 0.2 micron, low protein-binding 13 mm syringe-filter and stored at 4 °C. Method F. Conjugations were performed using a Tecan Freedom Evo robotic liquid handling system. The solution of antibody (10 mg/mL) was preheated to 37 °C and aliquoted to a heated 96 deep-well plate in amounts of 3 mg per well (0.3 mL) and kept at 37°C. A solution of Bond-Breaker™ tris(2- carboxyethyl)phosphine (TCEP) solution (1 mM, 0.051 mL/well) was added to antibodies, and the reaction mixture was kept at 37 °C for 75 minutes. The solution of reduced antibody was transferred to an unheated 96 deep-well plate. Corresponding solutions of synthons (5 mM, 0.024 mL in DMSO) were added to the wells with reduced antibodies and treated for 15 minutes. The reaction solutions were loaded onto a platform (8 x 12) of desalting columns (NAP5, washed with DPBS 4x before use), followed by DPBS (0.3 mL) and eluted with additional DPBS (0.8 mL). The purified ADC solutions were further aliquoted for analytics and stored at 4 °C. Method G. Conjugations were performed using a Tecan Freedom Evo robotic liquid handling system. The solution of antibody (10 mg/mL) was preheated to 37 °C and aliquoted onto a heated 96 deep- well plate in amounts of 3 mg per well (0.3 mL) and kept at 37 C. A solution of Bond-Breaker™ tris(2-carboxyethyl)phosphine (TCEP) solution (1 mM, 0.051 mL/well) was added to antibodies, and the reaction mixture was kept at 37 °C for 75 minutes. The solutions of reduced antibody were transferred to an unheated 96 deep-well plate. Corresponding solutions of synthons (5 mM, 0.024 mL/well in DMSO) were added to the wells with reduced antibodies followed by addition of boric buffer (pH=8, 0.03 mL/well) and treated for 3 days. The reaction solutions were loaded onto a platform (8 x 12) of desalting columns (NAP5, washed with DPBS 4x before use), followed by DPBS (0.3 mL) and eluted with additional DPBS (0.8 mL). The purified ADC solutions were further aliquoted for analytics and stored at 4 °C. Method H. A solution of Bond-Breaker™ tris(2-carboxyethyl)phosphine (TCEP) solution (10 mM, 017 mL) was added to the solution of antibody (10 mg/mL 10 mL) at room temperature The reaction mixture was heated to 37 °C for 75 minutes. The solution of synthon (10 mM, 0.40 mL in DMSO) was added to a solution of reduced antibody cooled to room temperature. The reaction solution was let to stand for 30 minutes at room temperature. The solution of ADC was treated with saturated ammonium sulfate solution (~2– 2.5 mL) until a slightly cloudy solution formed. This solution was loaded onto butyl sepharose column (5 mL of butyl sepharose) equilibrated with 30% phase B in phase A (phase A: 1.5 M ammonium sulphate, 25 mM phosphate; phase B: 25 mM phosphate, 25% isopropanol v/v). Individual fractions with DAR2 (also referred to as“E2”) and DAR4 (also referred to as“E4”) eluted upon applying gradient A/B up to 75% phase B. Each ADC solution was concentrated and buffer switched using centrifuge concentrators or TFF for larger scales. The purified ADC solutions were filtered through a 0.2 micron, low protein-binding 13 mm syringe- filter and stored at 4 °C. Method I. A solution of Bond-Breaker™ tris(2-carboxyethyl)phosphine (TCEP) solution (10 mM, 0.17 mL) was added to the solution of antibody (10 mg/mL, 10 mL) at room temperature. The reaction mixture was heated to 37 °C for 75 minutes. The solution of synthon (10 mM, 0.40 mL in DMSO) was added to a solution of reduced antibody cooled to room temperature. The reaction solution was let to stand for 30 minutes at room temperature. The solution of ADC was treated with saturated ammonium sulfate solution (~2– 2.5 mL) until a slightly cloudy solution formed. This solution was loaded onto a butyl sepharose column (5 mL of butyl sepharose) equilibrated with 30% phase B in Phase A (phase A: 1.5 M ammonium sulphate, 25 mM phosphate; phase B: 25 mM phosphate, 25% isopropanol v/v). Individual fractions with DAR2 (also referred to as“E2”) and DAR 4 (also referred to as“E4”) eluted upon applying a gradient A/B up to 75% phase B. Each ADC solution was concentrated and buffer switched using centrifuge concentrators or TFF for larger scales. The ADC solutions were treated with boric buffer (0.1 mL, 1M, pH8). The reaction solution was let stand for 3 days at room temperature, then loaded onto a desalting column (PD10, washed with DPBS 3x5mL before use), followed by DPBS (1.6 mL) and eluted with additional DPBS (3 mL). The purified ADC solution was filtered through a 0.2 micron, low protein-binding 13 mm syringe-filter and stored at 4 °C. DAR and aggregation of ADCs
The DAR and percentage aggregation of ADCs synthesized were determined by LC-MS and size exclusion chromatography (SEC), respectively. LC-MS general methodology
LC-MS analysis was performed using an Agilent 1100 HPLC system interfaced to an Agilent LC/MSD TOF 6220 ESI mass spectrometer. The ADC was reduced with 5 mM (final concentration) Bond-Breaker® TCEP solution (Thermo Scientific, Rockford, IL), loaded onto a Protein Microtrap (Michrom Bioresorces, Auburn, CA) desalting cartridge, and eluted with a gradient of 10% B to 75% B in 0.2 minutes at ambient temperature. Mobile phase A was H20 with 0.1% formic acid (FA), mobile phase B was acetonitrile with 0.1% FA, and the flow rate was 0.2 mL/min. Electrospray- ionization time-of-flight mass spectra of the co-eluting light and heavy chains were acquired using Agilent MassHunterTM acquisition software. The extracted intensity vs. m/z spectrum was deconvoluted using the Maximum Entropy feature of MassHunter software to determine the mass of each reduced antibody fragment. DAR was calculated from the deconvoluted spectrum by summing intensities of the naked and modified peaks for the light chain and heavy chain, normalized by multiplying intensity by the number of drugs attached. The summed, normalized intensities were divided by the sum of the intensities, and the summing results for two light chains and two heavy chains produced a final average DAR value for the full ADC.
Thiosuccinimide hydrolysis of a bioconjugate can be monitored by electrospray mass spectrometry, since the addition of water to the conjugate results in an increase of 18 Daltons to the observable molecular weight of the conjugate. When a conjugate is prepared by fully reducing the interchain disulfides of a human IgG1 antibody and conjugating the maleimide derivative to each of the resulting cysteines, each light chain of the antibody will contain a single maleimide modification and each heavy chain will contain three maleimide modifications, as described in Figure 2. Upon complete hydrolysis of the resulting thiosuccinimides, the mass of the light chain will therefore increase by 18 Daltons, while the mass of each heavy chain will increase by 54 Daltons. This is illustrated in Figure 5 with the conjugation and subsequent hydrolysis of an exemplary maleimide drug-linker (synthon TX, molecular weight 1736 Da) to the fully reduced huAb13v1 antibody. Size exclusion chromatography general methodology
Size exclusion chromatography was performed using a Shodex KW802.5 column in 0.2 M potassium phosphate pH 6.2 with 0.25 mM potassium chloride and 15% IPA at a flow rate of 0.75 ml/min. The peak area absorbance at 280 nm was determined for each of the high molecular weight and monomeric eluents by integration of the area under the curve. The % aggregate fraction of the conjugate sample was determined by dividing the peak area absorbance at 280 nM for the high molecular weight eluent by the sum of the peak area absorbances at 280 nM of the high molecular weight and monomeric eluents multiplied by 100%. In vitro cell viability assay methods
The tumor cell lines HCC38 (breast cancer), NCI-H1650 (NSCL), and NCI-H847 (small cell lung cancer cell line) were obtained from American Type Culture Collection (ATCC). Cells were grown in 96-well culture plates using recommended growth media overnight at a density of 5 x 103 (HCC38) or 20 x 103 (NCI-H847) or 40 x 103 (NCI-H1650) per well. The following day, treatments were added in fresh media to triplicate wells. Cellular viability was determined 5 days later using the CellTiter-Glo Luminescent Cell Viability Assay kit (Promega), as directed in the manufacturer’s protocol. Cell viability was assessed as percentage of control untreated cells. Example 8 In vivo Efficacy of Anti-B7-H3 Antibody Drug Conjugates
Of the nine chimeric antibodies tested in vitro conjagted to CZ synthons, four showed subnanomolar cytotoxicity (Table 6). chAb3-CZ, chAb18-CZ, and chAb13-CZ achieved DARS ranging from 2.6 to 4.2 (see Table 7) and were assessed for anti-tumor activity in a mouse small cell lung cancer cell line xenograft model NCI-H146, of human origin, using the methods described below. Antibody MSL109 (an IgG1 antibody that binds to cytomegalovirus (CMV) glycoprotein H) was used as a control, both as a naked antibody and as an ADC (conjugated to the same synthon (CZ) as the chAb3, chAb18, and chAb13 antibodies). MSL109 is a monoclonal antibody to CMV early protein and is an isotype matched non-targeting control. The methods of this xenograft assay are described below. The results are presented in Table 7. The results show that each of the anti-B7-H3 Bcl-xL inhibiting ADCs were able to significantly inhibit tumor growth relative to the naked antibody control (MSL109) or non-target specific Bcl-xL ADC control (MSL109-CZ).
Table 7: In vivo efficacy of chimeric anti-B7-H3 antibody as Bcl-xL drug conjugates
Figure imgf000490_0001
Evaluation of efficacy in xenograft models methods
NCI-H146 cells, NCI-1650 cells and EBC-1 cells were obtained from the American Type Culture Collection (ATCC, Manassas, VA). The cells were cultured as monolayers in RPMI-1640 (NCI-H146, NCI-H1650) or MEM (EBC-1) culture media (Invitrogen, Carlsbad, CA) that was supplemented with 10 % Fetal Bovine Serum (FBS, Hyclone, Logan, UT). To generate xenografts, 5x106 viable cells were inoculated subcutaneously into the right flank of immune deficient female SCID/bg mice (Charles River Laboratories, Wilmington, MA) respectively. The injection volume was 0.2 mL and composed of a 1:1 mixture of S MEM and Matrigel (BD, Franklin Lakes, NJ). Tumors were size matched at approximately 200 mm3. Antibodies and conjugates were formulated in 0.9% sodium chloride for injection and injected intraperitoneally. Injection volume did not exceed 200 µL. Therapy began within 24 hours after size matching of the tumors. Mice weighed approximately 22 g at the onset of therapy. Tumor volume was estimated two to three times weekly. Measurements of the length (L) and width (W) of the tumor were taken via electronic caliper and the volume was calculated according to the following equation: V = L x W2/2. Mice were euthanized when tumor volume reached 3,000 mm3 or skin ulcerations occurred. Eight mice were housed per cage. Food and water were available ad libitum. Mice were acclimated to the animal facilities for a period of at least one week prior to commencement of experiments. Animals were tested in the light phase of a 12-hour light: 12-hour dark schedule (lights on at 06:00 hours). As described above, human IgG control antibody (MSL109) was used as a negative control agent.
To refer to efficacy of therapeutic agents, parameters of amplitude (TGImax), durability (TGD) of therapeutic response are used. TGImax is the maximum tumor growth inhibition during the experiment. Tumor growth inhibition is calculated by 100*(1-Tv/Cv) where Tv and Cv are the mean tumor volumes of the treated and control groups, respectively. TGD or tumor growth delay is the extended time of a treated tumor needed to reach a volume of 1 cm3 relative to the control group. TGD is calculated by 100*(Tt/Ct-1) where Tt and Ct are the median time periods to reach 1 cm3 of the treated and control groups, respectively. Example 9: Humanization of Anti-B7-H3 Antibody chAb18
Anti-B7-H3 chimeric antibody chAb18 was selected for humanization based on its binding characteristics and favorable properties as an ADC, including its properties when conjugated to a Bcl- xL inhibitor (described above as exemplary conjugate CZ).
Humanized antibodies were generated based on the variable heavy (VH) and variable light (VL) CDR sequences of chAb18. Specifically, human germline sequences were selected for constructing CDR-grafted, humanized chAb18 antibodies, where the CDR domains of the VH and VL chains were grafted onto different human heavy and light chain acceptor sequences. Based on the alignments with the VH and VL sequences of monoclonal antibody chAb18, the following human sequences were selected as acceptors:
• IGHV1-69*06 and IGHJ6*01 for constructing heavy chain acceptor sequences
• IGKV1-9*01 and IGKJ2*01 for constructing light chain acceptor sequences
• IGKV6-21*01and IGKJ2*01 as backup acceptor for constructing light chain
Thus, the VH and VL CDRs of chAb18 were grafted into said acceptor sequences.
To generate humanized antibodies, framework back-mutations were identified and introduced into the CDR-grafted antibody sequences by de novo synthesis of the variable domain, or mutagenic oligonucleotide primers and polymerase chain reactions, or both by methods well known in the art. Different combinations of back mutations and other mutations were constructed for each of the CDR- grafts as described below. Residue numbers for these mutations are based on the Kabat numbering system.
For heavy chains huAb18VH.1, one or more of the following Vernier and VH/VL interfacing residues were back mutated as follows: L46P, L47W, G64V, F71H. Additional mutations include the following: Q1E, N60A, K64Q, D65G. For light chains huAb18VL.1, one or more of the following Vernier and VH/VL interfacing residues were back mutated as follows: A43S, L46P, L47W, G64V, G66V, F71H. For light chains huAb18VL.2, one or more of the following Vernier and VH/VL interfacing residues were back mutated as follows: L46P, L47W, K49Y, G64V, G66V, F71H.
The variable region and CDR amino acid sequences of the humanized antibodies are described in Table 8 below. TABLE 8: VH and VL amino acid sequences of humanized versions of chAb18
Figure imgf000492_0001
Figure imgf000493_0001
Figure imgf000494_0001
Humanized variable regions of the murine monoclonal Ab18 (described above) were cloned into IgG expression vectors for functional characterization:
• Humanized Ab18VH.1 (huAb18VH.1) is a CDR-grafted, humanized Ab18 VH containing IGHV1-69*06 and IGHJ6*01 framework sequences. It also contains a Q1E change to prevent pyroglutamate formation. The variable and CDR sequences of huAb18VH.1 are described in Table 8.
• Humanized Ab18VH1.a (huAb18VH.1a) is a humanized design based on huAb18VH.1 and contains 4 proposed framework back-mutations: M48I, V67T, L69I, K73R. The variable and CDR sequences of huAb18VH.1a are described in Table 8.
• Humanized Ab18VH1.b (huAb18VH.1b) is a humanized design based on huAb18VH.1 and huAb18VH.1a and contains 1 proposed framework back-mutation L69I and 3 HCDR2 germlining changes N60A, K64Q, D65G. The variable and CDR sequences of
huAb18VH.1b are described in Table 8.
• Humanized Ab18VL.1 (huAb18VL.1) is a CDR-grafted, humanized Ab18 VL containing IGKV1-9*01 and IGKJ2*01 framework sequences. The variable and CDR sequences of huAb18VL.1 are described in Table 8.
• Humanized Ab18VL.1a (huAb18VL.1a) is a humanized design based on huAb18VL.1 and contains 6 proposed framework back-mutations: A43S, L46P, L47W, G64V, G66V, F71H. The variable and CDR sequences of huAb18VL.11 are described in Table 8.
• Humanized Ab18VL.1b (huAb18VL.1b) is a humanized design based on huAb18VL.1 and huAb18VL.1a contains 4 proposed framework back-mutations: L46P, L47W, G64V, F71H. The variable and CDR sequences of huAb18VL.1b are described in Table 8.
• Humanized Ab18VL.2 (huAb18VL.2) is a CDR-grafted, humanized Ab18 VL containing IGKV6-21*01 and IGKJ2*01 framework sequences. The variable and CDR sequences of huAb18VL.2 are described in Table 8.
• Humanized Ab18VL.2a (huAb18VL.2a) is a humanized design based on huAb18VL.2 and contains 6 proposed framework back-mutations: L46P, L47W, K49Y, G64V, G66V, F71H. The variable and CDR sequences of huAb18VL.2a are described in Table 8.
Thus, the humanization of chAb18 resulted in 10 humanized antibodies, including huAb18v1, huAb18v2, huAb18v3, huAb18v4, huAb18v5, huAb18v6, huAb18v7, huAb18v8, huAb18v9, and huAb18v10. The variable and heavy light chains for each of these humanized versions of Ab18 are provided below:
Table 9: Anti-B7-H3 Ab18 humanized antibodies
Figure imgf000495_0001
Example 10: In vitro Characterization of Anti-B7-H3 chAb18 Humanized Variants
The humanization of chAb18 generated 10 variants (described above in Table 9) that retained binding to human and cyno B7-H3 as assessed by FACS (the method of which is described above in Example 6). These variants were further characterized for binding by SPR and were successfully conjugated to the Bcl-xL inhibitor synthon CZ using Method A (described above) and assessed for cell cytotoxicity as described in Example 7. Table 10 summarizes the in vitro characteristics of the various humanized Ab18 variants. The parental chAb18 from which the variants were derived was also tested as a comparator. All humanized variants had similar binding properties as assessed by Biacore, and retained binding activity to cell surface expressed as conjugates with the CZ synthon. The cytotoxicity of all of the variants as CZ synthons were similar to the chAb18 from which they were derived.
TABLE 10: In vitro characterization of humanized anti-B7-H3 Ab18 variants
Figure imgf000495_0002
Humanized chAb18 variants were conjugated to the CZ synthon and tested for cytotoxicity in HCC38 cell line. As described in Table 10, most humanized antibodies showed potent cytotoxicity, similar to those observed with control antibody chAb18. Example 11: In Vivo Efficacy of Humanized Ab18 Variants as Bcl-xL Inhibitor ADCs
Six of the humanized chAb18 variants were selected based on in vitro cytotoxicity results described in Example 10. Specifically, antibodies huAb18v1, huAb18v3, huAb18v4, huAb18v6, huAb18v7, and huAb18v9 were each conjugated to the CZ synthon (to form an anti-B7-H3 CZ ADC) for evaluation in an in vivo xenograft model of small cell lung cancer (using NCI-H146 cells), as described in Example 8. Single dose treatment of the tumor bearing mice resulted in tumor growth inhibition and tumor growth delay and the results are summarized in Table 11. Ab095 was used as a negative control for the effect of administering IgG, as it is an isotype matched non-target specific antibody raised against tetanus toxoid. See Larrick et al., 1992, Immunological Reviews 69-85. Mice were administered 6 mg/kg of the ADC intraperitoneally QDx1.
Table 11: In vivo efficacy of anti-B7-H3 ADCS (humanized chAb18-CZ variants)
Figure imgf000496_0001
As described in Table 11, each of the tested humanized antibodies was able to inhibit tumor growth in the mouse xenograft model. Example 12: Humanization of Anti-B7-H3 Antibody chAb3
Anti-B7-H3 chimeric antibody chAb3 was selected for humanization based on its favorable properties as a Bcl-xL inhibiting (Bcl-xLi) conjugate. Humanized antibodies were generated based on the variable heavy (VH) and variable light (VL) CDR sequences of chAB3. Specifically, human germline sequences were selected for constructing CDR-grafted, humanized chAb3 antibodies where the CDR domains of the VH and VL chains of chAb3 were grafted onto different human heavy and light chain acceptor sequences. Based on the alignments with the VH and VL sequences of monoclonal antibody chAb3 the following human sequences were selected as acceptors:
• IGHV1-69*06 and IGHJ6*01 for constructing heavy chain acceptor sequences
• IGKV2-28*01 and IGKJ4*01 for constructing light chain acceptor sequences By grafting the corresponding VH and VL CDRs of chAb3 into said acceptor sequences, CDR-grafted, humanized, and modified VH and VL sequences were prepared. To generate humanized antibodies with potential framework back-mutations, the mutations were identified and introduced into the CDR-grafted antibody sequences by de novo synthesis of the variable domain, or mutagenic oligonucleotide primers and polymerase chain reactions, or both. Different combinations of back mutations and other mutations are constructed for each of the CDR-grafts as follows. Residue numbers for these mutations are based on the Kabat numbering system.
The amino acid sequences of the various humanized heavy and light chain variable regions are described below in Table 12.
For heavy chains huAb3VH.1, one or more of the following Vernier and VH/VL interfacing residues were back mutated as follows: M48I, V67A, I69L, A71V, K73R, M80V, Y91F, R94G. For light chains huAb31 VL.1, one or more of the following Vernier and VH/VL interfacing residues were back mutated as follows: I2V, Y87F.
The following humanized variable regions of the murine monoclonal chAb3 antibody were cloned into IgG expression vectors for functional characterization:
• Humanized Ab3 VH.1 (huAb3VH.1) is a CDR-grafted, humanized Ab3 VH containing
IGHV1-69*06 and IGHJ6*01 framework sequences. It also contains a Q1E change to prevent pyroglutamate formation.
• Humanized Ab3 VH.1a (huAb3VH.1a) is a humanized design based on huAb3VH.1 and contains 8 proposed framework back-mutations: M48I, V67A, I69L, A71V, K73R, M80V, Y91F, R94G.
• Humanized Ab3 VH.1b (huAb3VH.1b) is a humanized design between huAb3VH.1 and huAb3VH.1a and contains 6 proposed framework back-mutations: M48I, V67A, I69L, A71V, K73R, R94G.
• Humanized Ab3 VL.1 (huAb3VL.1) is a CDR-grafted, humanized Ab3 VL containing
IGKV2-28*01 and IGKJ4*01 framework sequences.
• Humanized Ab3 VL.1a (huAb3VL.1a is a humanized design based on huAb3VL.1 and
contains 2 proposed framework back-mutations: I2V, Y87F.
• Humanized Ab3 VL.1b (huAb3VL.1b) is a humanized design contains only 1 proposed
framework back-mutations: I2V.
Further to the above, exemplary framework sequences are described below: IGHV1-69*06_IGHJ6
QVQLVQSGAEVKKPGSSVKVSCKASggtfssyaisWVRQAPGQGLEWMGgiipifgtanyaqkfqgRVTI TADKSTSTAYMELSSLRSEDTAVYYCARxxxxxxxxWGQGTTVTVSS (SEQ ID NO: 174);
where xxxxxxxx represents the CDR-H3 region. IGKV2-28*01_IGKJ4
DIVMTQSPLSLPVTPGEPASISCrssqsllhsngynyldWYLQKPGQSPQLLIYlgsnrasGVPDRFSGSGS GTDFTLKISRVEAEDVGVYYCxxxxxxxxxFGGGTKVEIK (SEQ ID NO: 175);
where xxxxxxxxx represents the CDR-L3 region. The variable region and CDR amino acid sequences of the foregoing humanized antibodies are described in Table 12 below.
Figure imgf000498_0001
Figure imgf000499_0002
The humanization of chAb3 resulted in 6 humanized antibodies, including huAb3v1, huAb3v2, huAb3v3, huAb3v4, huAb18v5, and huAb3v6. The variable and heavy light chains for each of these humanized versions of Ab18 are provided below in Table 13.
Table 13: Humanized Ab3 antibodies
Figure imgf000499_0001
Example 13: In vitro characterization of chAb3 humanized variants
The humanization of chAb3 generated 6 variants (described in Table 13) that retained binding to human B7-H3 as assessed by FACS (as described in Example 6). These variants were further characterized for binding by SPR and as ADCs conjugated to the Bcl-xL inhibitor synthon (linker warhead) CZ. The humanized Ab3 antibodies were also assessed for cell cytotoxicity (using the assay described above in Example 7). Table 14 summarizes in vitro characteristics of chAb3 humanized variants. An ADC comprising chAb3 conjugated to synthon CZ was used as a control. TABLE 14: In vitro characterization of humanized variants of chAb3
Figure imgf000500_0001
Example 14: In vivo Efficacy of chAb3 Humanized Variants as Bcl-xL ADCs
Two of the humanized variants (huAb3v2 and huAb3v6) were selected based on in vitro cytotoxicity as CZ conjugates for evaluation in an in vivo murine xenograft model of small cell lung cancer cells (NCI-H146 cells) as described in materials and methods in Example 8. Single dose treatment of tumor bearing mice resulted in tumor growth inhibition and tumor growth delay for both humanized antibodies conjugated to an exemplary Bcl-xL inhibitor, and the results are summarized in Table 15.
Table 15: In vivo efficacy of humanized chAb3-CZ variants
Figure imgf000500_0002
Example 15: Modifications of the CDRs of Humanized Variant Antibody huAb3v2
huAb3v2 showed favorable binding and cell killing properties. An examination of the variable region amino acid sequences of huAb3v2, however, revealed potential deamidation and/or isomerization sites.
The amino acid sequences of huAb3 variable regions are described below, including the light chain (huAb3VL1) and the heavy chain (huAb3VH1b). The potential deamidation and/or isomerization sites in CDRs of the VH (CDR2 at amino acids“ds” and VL (CDR1 at amino acids “ng”) are italicized and were thus engineered to improve antibody manufacturing. The CDRs are described in lower case letters in the sequences below.
To make huAb3v2 variants lacking these potential deamidation and/or isomerization sites, each of the amino acids indicated below (x and z; representing the potential sites in the CDR1 of the VL and the CDR2 of the VH) were mutagenized. The resulting 30 VL variants were paired with the original huAb3v2 VH and tested for binding. The resulting 29 VH variants were paired with the original huAb3v2 VL and tested for binding. Successful VH variants were combined and tested with productive VL variants harboring changes in LCDR1 to make the final humanized variants lacking the potential deamidation and/or isomerization sites in CDRs. The amino acid sequences of the variants are provided in Table 16 below. The full length amino acid sequences of the heavy chain and light chain of the huAb3v2 variant, huAb3v2.5 are provided in SEQ ID NOs: 170 and 171, respectively. The full length amino acid sequences of the heavy chain and light chain of the huAb3v2 variant, huAb3v2.6 are provided in SEQ ID NOs: 172 and 173, respectively. huAb3 VL1
DIVMTQSPLSLPVTPGEPASISCrssqslvhsngdtylrWYLQKPGQSPQLLIYkvsnrfsGVPDRFSGSGS GTDFTLKISRVEAEDVGVYYCsqsthvpytFGGGTKVEIK (SEQ ID NO: 128)
xg (15 variants) (SEQ ID NO: 178)
nz (15 variants) (SEQ ID NO: 179)
huAb3 VH1b
EVQLVQSGAEVKKPGSSVKVSCKASgytfssywmhWVRQAPGQGLEWIGlihpdsgstnynemfknRAT LTVDRSTSTAYMELSSLRSEDTAVYYCAGggrlyfdyWGQGTTVTVSS (SEQ ID NO: 127)
(15 variants) xs (SEQ ID NO: 180)
(14 variants) dz (SEQ ID NO: 181)
where (for both the VL and VH),
x = All amino acids, except: M, C, N, D, or Q.
z = All amino acids, except: M, C, G, S, N, or P.
Proposed framework back mutations are underlined (see Example 12).
Table 16: Variable region sequences of huAb3v2 antibody variants
Figure imgf000501_0001
Figure imgf000502_0001
Figure imgf000503_0001
Figure imgf000504_0001
Figure imgf000505_0001
Example 16: In vitro characterization of huAb3v2 Variants
Removal of potential deamidation and/or isomerization sites (described in Example 15) generated only 6 variants that retained binding to both human and cyno B7-H3 exogenously expressed on mouse 3T12 fibroblasts as assessed by FACS (as described in the methods of Example 6).
These new anti-B7-H3 antibodies were further characterized for binding by SPR and conjugated to the Bcl-xLi synthon CZ and assessed for cell cytotoxicity (using the methods described in Example 7). Table 17 provides in vitro characteristics of six huAb3v2 humanized variants.
Table 17: In vitro characterization of humanized huAb3v2 variants, including naked anitbodies and ADCs
Figure imgf000506_0001
As described in Table 17, the results showed that all six huAb3v2 variants had similar binding properties to cells expressing human or cynoB7-H3 as compared to the parental huAb3v2. Of the six huAb3v2 variants, four antibodies (huAb3v2.5, huAb3v2.6, huAb3v2.8, huAb3v2.9) showed potent cytotoxicity in H847 cells when conjugated to exemplary Bcl-xLi synthon CZ. Example 17: Humanization of Anti-B7-H3 Antibody chAb13
The anti-B7-H3 chimeric antibody chAb13 was selected for humanization based on its binding characteristics and favorable properties as an ADC (conjugated to a Bcl-xL inhibitor).
Prior to humanization, chAb13 was modified in order to minimize potential deamidation in the light chain CDR3 (QQYNSYPFT (SEQ ID NO:182); potential deamidation site is indicated as residues“NS” (italicized)). Point mutations in the amino acid position corresponding to“N” and/or “S” within the light chain CDR3 of chAb13 were introduced, resulting in 30 variants. Antibodies containing these CDR3 light chain variants were then screened for their ability to retain the binding characteristics of chAb13. Variants comprising a CDR3 having a tryptophan (W) point mutation instead of the serine“S” in the“NS” motif (i.e., QQYNWYPFT (SEQ ID NO: 39)) retained the binding features of the parent chAb13 antibody. The substitution of the S residue with a W residue within the CDR3was surprising given the structural differences between serine and tryptophan as well as the significant role the CDR3 plays in antigen binding.
Humanized antibodies were generated based on the variable heavy (VH) and variable light (VL) CDR sequences of chAb13, including the“NW” light chain CDR3. Specifically, human germline sequences were selected for constructing CDR-grafted, humanized chAb13 antibodies, where the CDR domains of the VH and VL chains were grafted onto different human heavy and light chain acceptor sequences. Based on the alignments with the VH and VL sequences of monoclonal antibody chAb13, the following human sequences were selected as acceptors:
• IGHV4-b*01(0-1) and IGHJ6*01 for constructing heavy chain acceptor sequences • IGKV1-39*01 and IGKJ2*01 for constructing light chain acceptor sequences IGHV4-b_IGHJ6
QVQLQESGPGLVKPSETLSLTCAVSgysissgyywgWIRQPPGKGLEWIGsiyhsgstyynpslksRVTISV DTSKNQFSLKLSSVTAADTAVYYCARxxxxxxxWGQGTTVTVSS (SEQ ID NO: 176);
where xxxxxxx represents the CDR-H3 region.
IGKV1-39_IGKJ2
DIQMTQSPSSLSASVGDRVTITCrasqsissylnWYQQKPGKAPKLLIYaasslqsGVPSRFSGSGSGTD FTLTISSLQPEDFATYYCxxxxxxxxxFGQGTKLEIK (SEQ ID NO: 177);
where xxxxxxxxx represents the CDR-L3 region. By grafting the“NW” light chain CDR3 and the remaining five corresponding VH and VL CDRs of chAb13 into said acceptor sequences, CDR-grafted, humanized, and modified VH and VL sequences were prepared. To generate humanized antibodies with potential framework back- mutations, the mutations were identified and introduced into the CDR-grafted antibody sequences by de novo synthesis of the variable domain, or mutagenic oligonucleotide primers and polymerase chain reactions, or both by methods well known in the art. Different combinations of back mutations and other mutations were constructed for each of the CDR-grafts as follows. Residue numbers for these mutations are based on the Kabat numbering system.
The following humanized variable regions of the murine monoclonal chAb13 antibodies were cloned into IgG expression vectors for functional characterization:
• Humanized Ab13 VH.1 (huAb13VH.1) is a CDR-grafted, humanized Ab13 VH containing IGHV4-b*01(0-1) and IGHJ6*01 framework sequences. It also contains a Q1E change to prevent pyroglutamate formation.
• Humanized Ab13 VH.1 (huAb13 VH.1a) is a humanized design based on huAb13VH.1 and contains 9 proposed framework back-mutation(s): S25T, P40F, K43N, I48M, V67I, T68S, V71R, S79F, R94G. • Humanized Ab13 VH.1b (huAb13VH.1b) is an intermediate design between on huAb13VH.1 and huAb13VH.1a and contains 4 proposed framework back-mutation(s): K43N, I48M, V67I, V71R.
• Humanized Ab13 VL.1 (huAb13VL.1) is a CDR-grafted, humanized Ab13 VL containing IGKV1-39*01 and IGHJ6*01 framework sequences.
• Humanized Ab13 VL.1a (huAb13VL.1a) is a humanized design based on huAb13VL.1 and contains 4 proposed framework back-mutation(s): A43S, L46A, T85E, Y87F.
• Humanized Ab13 VL.1b (huAb13VL.1b) is an intermediate design between on huAb13VL.1 and huAb13VL.1a and contains 1 proposed framework back-mutation(s): Y87F.
The variable region and CDR amino acid sequences of the foregoing are described in Table 18 below.
Table 18: Amino acid variable region sequences of humanized Ab13
Figure imgf000508_0001
Figure imgf000509_0001
Figure imgf000510_0002
Example 18: Generation of huAb13 variants
The 3 VH and 3 VL region amino acid sequences of humanized Ab13 variants described in Table 18 were paired together to generate 9 huAb13 variants described in Table 19. The full length amino acid sequences of the heavy chain and light chain of the huAb13v1 variant, huAb13v1 are provided in SEQ ID NOs: 168 and 169, repectively. Table 19: Variable region sequences of engineered huAb13 variants
Figure imgf000510_0001
Figure imgf000511_0001
Figure imgf000512_0001
Figure imgf000513_0001
Figure imgf000514_0001
Figure imgf000515_0002
Example 19: Characterization of huAb13 VL.1a Humanized Variants
Nine huAb13 variants described in Examples 17 and 18 were generated and tested for binding to B7-H3 by FACS (according to methods described in Example 6). Six variants did not bind to human B7-H3. The remaining three variants were further characterized for binding by SPR and conjugated (via Method A) to the Bcl-xL inhibitor (specifically the linker warhead (or synthon) CZ) and assessed for cell cytotoxicity (according to methods described in Example 7). Table 20 provides the in vitro characteristics of these variants. Table 20: In vitro characterization of huAb13 VL.1a variants conjugated to payload CZ
Figure imgf000515_0001
Figure imgf000516_0002
HuAb13v1 was selected for further study due in part to its potent and superior cytotoxicity against H847 cells and similar binding characteristics as chAb13 from which it was derived. In contrast, huAb13v5 and huAb13v6 showed poor fit kinetics in Biacore experiments suggesting their binding properties are more divergent from the parental chAb13 than huAb13v1 and have reduced activity in the cell killing assay. Example 20: In vitro potency of Selected Humanized B7-H3 Antibodies with Exemplary Bcl-xL Inhibitor Linker Warheads (Synthons)
Humanized antibodies huAb13v1, huAb3v2.5 and huAb3v2.6 were selected to be conjugated with several Bcl-xL inhibitor payloads (or synthons) at a 3 mg scale using Methods A, E or G, as described in Example 7. The anti-tumor activity of these ADCs was tested in cytotoxicity assays using the NCI-H1650 non-small cell lung cancer cell line as described in Example 7. As control, the in vitro anti-tumor activity of ADCs comprising the non-targeting antibody MSL109 (a monoclonal antibody that binds to the CMV glycoprotein H conjugated to Bcl-xL inhibitor payloads (or synthons) was also evaluated. The results are described in Table 21. Table 21: In vitro tumor cell cytotoxicity of selected humanized B7-H3 ADCs with exemplary Bcl- xL inhibitor linker warheads (synthons)
Figure imgf000516_0001
Figure imgf000517_0001
Figure imgf000518_0003
As described in Table 21, Bcl-xLi ADCs comprising humanized anti-B7-H3 antibodies and specific synthons, e.g. synthon XW, were able to kill tumor cells in vitro. In contrast to the low anti- tumor activity exhibited by the ADCs comprising the non-targeting antibody MSL109 conjugated to a Bcl-xL inhibitor payload, the B7-H3-targeting ADCs exhibited greater tumor cell killing, which reflects the antigen-dependent delivery of the B7-H3-targeting ADCs to the B7-H3-expressing tumor cells.
In contrast to the low anti-tumor activity exhibited by the ADCs comprising the non-targeting antibody MSL109 conjugated to a Bcl-xL inhibitor payload, the B7-H3-targeting ADCs exhibited greater tumor cell killing, which reflects the antigen-dependent delivery of the B7-H3-targeting ADCs to the B7-H3-expressing tumor cells. Example 21: In vivo analysis of anti-B7-H3 ADCs
Humanized anti-B7-H3 antibodies huAb13v1, huAb3v2.5 and huAb3v2.6 were selected to be conjugated with several Bcl-xL inhibitor payloads (or synthons) and were evaluated in xenograft models of small cell lung cancer (H146) as conjugates using a number of Bcl-xL inhibitor payloads (or synthons) using the methods described in Examples 7 and 8. The results are summarized in Table 22 and Table 23. Table 22: In vivo efficacy of humanized anti-B7-H3 ADCs
Figure imgf000518_0001
a dose is given in mg/kg/day
Table 23: In vivo efficacy of humanized anti-B7-H3 ADCS
Figure imgf000518_0002
Figure imgf000519_0002
Humanized anti-B7-H3 antibody huAb13v1 was conjugated with the Bcl-xL inhibitor synthon WD and evaluated in a xenograft model of the B7-H3-positive small cell lung cancer (H1650) as conjugates using the methods described in Example 7 and Example 8. As control, the in vivo anti-tumor activity of a non-targeting IgG isotype matched antibody (AB095) was also evaluated. The results are summarized in Table 24. Table 24: In vivo efficacy of humanized anti-B7-H3 ADC huAb13v1-WD in H1650
Figure imgf000519_0001
In contrast to the lack of activity observed using the non-targeting IgG isotype-matched antibody Ab095, the B7-H3-targeting Bcl-xL ADCs exhibited tumor growth inhibition (TGI) and tumor growth delay (TGD), as shown in Tables 24 and 25, reflecting the antigen-dependent delivery of the B7-H3-targeting ADCs which deliver the Bcl-xL inhibitor to the B7-H3-expressing tumor cells in this xenograft mouse model. As an additional control, the in vivo anti-tumor activity of ADCs comprising the non-targeting antibody MSL109 conjugated with Bcl-xL inhibitor synthons was evaluated in the xenograft model of the B7-H3-positive small cell lung cancer (H1650). The activity of these ADCs was compared that of the non-targeting IgG isotype matched antibody, AB095, as control. As shown in Table 25, the ADCs comprising the non-targeting antibody MSL109 conjugated with Bcl-xL inhibitor synthons exhibited very modest tumor growth inhibition and low or no tumor growth delay. In contrast, the B7-H3-targeting Bcl-xL ADCs (as shown in Table 24) exhibited much greater tumor growth inhibition (TGI) and tumor growth delay (TGD), reflecting the antigen- dependent delivery of these ADCs to B7-H3-expressing cells in this mouse xenograft model. Table 25. In vivo efficacy of non-targeting (MSL109) Bcl-xL inhibiting ADCs in NCI-H1650 model of NSCLC
Figure imgf000520_0001
SEQUENCE SUMMARY
Figure imgf000521_0001
Figure imgf000522_0001
Figure imgf000523_0001
Figure imgf000524_0001
Figure imgf000525_0001
Figure imgf000526_0001
Figure imgf000527_0001
Figure imgf000528_0001
Figure imgf000529_0001
Figure imgf000530_0001
Figure imgf000531_0001
Figure imgf000532_0001
Figure imgf000533_0001
Figure imgf000534_0001
Figure imgf000535_0001
Figure imgf000536_0001
Figure imgf000537_0001
Figure imgf000538_0001
Figure imgf000539_0001
Figure imgf000540_0001
Figure imgf000541_0001
Figure imgf000542_0001
INCORPORATION BY REFERENCE
The contents of all references, patents, pending patent applications and published patents, cited throughout this application are hereby expressly incorporated by reference. EQUIVALENTS
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims

1. An isolated antibody, or antigen binding portion thereof, that binds to human B7-H3 (hB7- H3), wherein the antibody, or antigen binding portion thereof, comprises a heavy chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 12 and a light chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 15.
2. The antibody, or antigen binding portion thereof, of claim 1, wherein the antibody, or antigen binding portion thereof, comprises a heavy chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 140 and a light chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 7.
3. The antibody, or antigen binding portion thereof, of claim 1 or 2, wherein the antibody, or antigen binding portion thereof, comprises a heavy chain variable region comprising a CDR1 having the amino acid sequence of SEQ ID NO: 10 and a light chain variable region comprising a CDR1 having the amino acid sequence of either SEQ ID NO: 136 or 138.
4. An isolated antibody, or antigen binding portion thereof, that binds to human B7-H3, wherein the antibody, or antigen binding portion thereof, comprises a heavy chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 35 and a light chain variable region comprising a CDR3 having the amino acid sequence of SEQ ID NO: 39.
5. The antibody, or antigen binding portion thereof, of claim 4, wherein the antibody, or antigen binding portion thereof, comprises a heavy chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 34, and a light chain variable region comprising a CDR2 having the amino acid sequence of SEQ ID NO: 38.
6. The antibody, or antigen binding portion thereof, of claim 4 or 5, wherein the antibody, or antigen binding portion thereof, comprises a heavy chain variable region comprising a CDR1 having the amino acid sequence of SEQ ID NO: 33 and a light chain variable region comprising a CDR1 having the amino acid sequence of either SEQ ID NO: 37.
7. The antibody, or antigen binding portion thereof, of any one of the preceding claims, wherein the antibody, or antigen binding portion thereof, is an IgG isotype.
8. The antibody, or antigen binding portion thereof, of claim 7, wherein the antibody, or antigen binding portion thereof is an IgG1 or an IgG4 isotype
9. The antibody, or antigen binding portion thereof, of any one of the preceding claims, wherein the antibody, or antigen binding portion thereof, has a KD of 1.5 x 10-8 or less as determined by surface plasmon resonance.
10. An antibody, or antigen-binding portion thereof, that binds to hB7-H3, said antibody, or antigen-binding portion thereof, comprising a heavy chain variable region comprising either a CDR set of SEQ ID NOs: 10, 11, and 12, and a light chain variable region comprising a CDR set of SEQ ID NOs: 14, 7, and 15, or
a heavy chain variable region comprising a CDR set of SEQ ID NOs: 33, 34, and 35, and a light chain variable region comprising a CDR set of SEQ ID NOs: 37, 38, and 39.
11. The antibody, or antigen binding portion thereof, of claim 10, wherein the antibody, or
antigen binding portion thereof, is humanized.
12. The antibody, or antigen binding portion thereof, according to claim 11, further comprising a human acceptor framework.
13. The antibody, or antigen binding portion thereof, of claim 12, wherein said human acceptor framework comprises an amino acid sequence selected from the group consisting of SEQ ID Nos: 155, 156, 164, 165, 166, and 167.
14. The antibody, or antigen binding portion thereof, of claim 13, wherein said human acceptor framework comprises at least one framework region amino acid substitution.
15. The antibody, or antigen binding portion thereof, of claim 14, wherein the amino acid
sequence of the framework is at least 65% identical to the sequence of said human acceptor framework and comprises at least 70 amino acid residues identical to said human acceptor framework.
16. The antibody, or antigen binding portion thereof, of claim 14 or 15, wherein said human acceptor framework comprises at least one framework region amino acid substitution at a key residue, said key residue selected from the group consisting of:
a residue adjacent to a CDR;
a glycosylation site residue;
a rare residue;
a residue capable of interacting with human B7-H3;
a residue capable of interacting with a CDR; a canonical residue;
a contact residue between heavy chain variable region and light chain variable region;
a residue within a Vernier zone; and
a residue in a region that overlaps between a Chothia-defined variable heavy chain CDR1 and a Kabat-defined first heavy chain framework.
17. The antibody, or antigen binding portion thereof, of claim 16, wherein said key residue is selected from the group consisting of 48H, 67H, 69H, 71H, 73H, 94H, and 2L.
18. The antibody, or antigen binding portion thereof, of claim 17, wherein the key residue
substitution is in the variable heavy chain region and is selected from the group consisting of M48I, V67A, I69L, A71V, K73R, and R94G.
19. The antibody, or antigen binding portion thereof, of claim 17 or 18, wherein the key residue substitution is in the variable light chain region and is I2V.
20. An antibody, or antigen-binding portion thereof, that binds to hB7-H3 comprising a heavy chain variable region comprising a CDR set of SEQ ID NOs: 25, 26, and 27, and a light chain variable region comprising a CDR set of SEQ ID NOs: 29, 30, and 31.
21. The antibody, or antigen binding portion thereof, of claim 20, wherein the antibody, or
antigen binding portion thereof, is humanized.
22. The antibody, or antigen binding portion thereof, according to claim 21, further comprising a human acceptor framework.
23. The antibody, or antigen binding portion thereof, of claim 22, wherein said human acceptor framework comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 155 to 158.
24. The antibody, or antigen binding portion thereof, of claim 22 or 23, wherein said human acceptor framework comprises at least one framework region amino acid substitution.
25. The antibody, or antigen binding portion thereof, of claim 24, wherein the amino acid
sequence of the framework is at least 65% identical to the sequence of said human acceptor framework and comprises at least 70 amino acid residues identical to said human acceptor framework
26. The antibody, or antigen binding portion thereof, of claim 24 or 25, wherein said human acceptor framework comprises at least one framework region amino acid substitution at a key residue, said key residue selected from the group consisting of:
a residue adjacent to a CDR;
a glycosylation site residue;
a rare residue;
a residue capable of interacting with human B7-H3;
a residue capable of interacting with a CDR;
a canonical residue;
a contact residue between heavy chain variable region and light chain variable region;
a residue within a Vernier zone; and
a residue in a region that overlaps between a Chothia-defined variable heavy chain CDR1 and a Kabat-defined first heavy chain framework.
27. The antibody, or antigen binding portion thereof, of claim 26, wherein said key residue is selected from the group consisting of 69H, 46L, 47L, 64L, and 71L.
28. The antibody, or antigen binding portion thereof, of claim 27, wherein the key residue
substitution is in the variable heavy chain region and is L69I.
29. The antibody, or antigen binding portion thereof, of claim 27 or 28, wherein the key residue substitution is in the variable light chain region and is selected from the group consisting of L46P, L47W, G64V, and F71H.
30. An anti-hB7-H3 antibody, or antigen-binding portion thereof, comprising a heavy chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 10, a heavy chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 140, a heavy chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 12, a light chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 136 or 138, a light chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 7, and a light chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 15.
31. An anti-hB7-H3 antibody, or antigen-binding portion thereof, comprising a heavy chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 33, a heavy chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 34, a heavy chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 35, a light chain CDR1 comprising an amino acid sequence as set forth in SEQ ID NO: 37 a light chain CDR2 comprising an amino acid sequence as set forth in SEQ ID NO: 38, and a light chain CDR3 comprising an amino acid sequence as set forth in SEQ ID NO: 39.
32. An anti-hB7-H3 antibody, or antigen-binding portion thereof, comprising a heavy chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 139 and a light chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 135.
33. An anti-hB7-H3 antibody, or antigen-binding portion thereof, comprising a heavy chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 139, and/or a light chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 135.
34. An anti-hB7-H3 antibody, or antigen-binding portion thereof, comprising a heavy chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 139 and a light chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 137.
35. An anti-hB7-H3 antibody, or antigen-binding portion thereof, comprising a heavy chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 139, and/or a light chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 137.
36. An anti-hB7-H3 antibody, or antigen-binding portion thereof, comprising a heavy chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 147 and a light chain variable domain comprising an amino acid sequence set forth in SEQ ID NO: 144.
37. An anti-hB7-H3 antibody, or antigen-binding portion thereof, comprising a heavy chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 147, and/or a light chain comprising an amino acid sequence having at least 90%, 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 144.
38. The antibody, or antigen-binding portion thereof, of any one of the preceding claims, wherein the antibody, or antigen binding portion thereof, binds cyno B7-H3.
39. The antibody, or antigen-binding portion thereof, of any one of the preceding claims, wherein the antibody, or antigen binding portion thereof, has a dissociation constant (KD) to hB7-H3 selected from the group consisting of: at most about 10-7 M; at most about 10-8 M; at most about 10-9 M; at most about 10-10 M; at most about 10-11 M; at most about 10-12 M; and at most 10-13 M.
40. The antibody, or antigen-binding portion thereof, of any one of the preceding claims, wherein the antibody, or antigen binding portion thereof, comprises a heavy chain immunoglobulin constant domain of a human IgM constant domain, a human IgG1 constant domain, a human IgG2 constant domain, a human IgG3 constant domain, a human IgG4 constant domain, a human IgA constant domain, or a human IgE constant domain and/or comprises a kappa light chain.
41. The antibody of any one of the preceding claims, which is an IgG1 antibody having four polypeptide chains, two heavy chains and two light chains.
42. The antibody, or antigen-binding portion thereof, of claim 41, wherein the human IgG1
constant domain comprises an amino acid sequence of SEQ ID NO: 159 or SEQ ID NO: 160.
43. An anti-hB7-H3 antibody comprising a sequence set selected from the group consisting of a) a heavy chain comprising the amino acid sequence of SEQ ID NO: 168 and a light chain comprising the amino acid sequence of SEQ ID NO: 169;
b) a heavy chain comprising the amino acid sequence of SEQ ID NO: 170 and a light chain comprising the amino acid sequence of SEQ ID NO: 171; and
c) a heavy chain comprising the amino acid sequence of SEQ ID NO: 172 and a light chain comprising the amino acid sequence of SEQ ID NO: 173.
44. An anti-hB7-H3 antibody, or antigen-binding portion thereof, that competes with the
antibody, or antigen binding portion thereof, of any one of the preceding claims.
45. A pharmaceutical composition comprising the anti-hB7-H3 antibody, or antigen binding portion thereof, of any one of claims 1-44, and a pharmaceutically acceptable carrier.
46. An anti-hB7-H3 Antibody Drug Conjugate (ADC) comprising an anti-hB7-H3 antibody of any one of claims 1-44 conjugated to a drug via a linker.
47. The ADC of claim 46, wherein the drug is an auristatin or a pyrrolobenzodiazepine (PBD).
48. The ADC of claim 46, wherein the drug is a Bcl-xL inhibitor.
49. An anti-hB7-H3 antibody drug conjugate (ADC) comprising a drug linked to an anti-human B7-H3 (hB7-H3) antibody by way of a linker, wherein the drug is a Bcl-xL inhibitor according to structural formula (IIa) or (IIb):
Figure imgf000549_0001
Figure imgf000549_0002
wherein:
Ar1 is selected from
Figure imgf000549_0003
Figure imgf000549_0004
and is optionally substituted with one or more substituents independently selected from halo, hydroxy, nitro, lower alkyl, lower heteroalkyl, C1-4alkoxy, amino, cyano and halomethyl;
Figure imgf000550_0001
Figure imgf000550_0002
is optionally substituted with one or more substituents independently selected from halo, hydroxy, nitro, lower alkyl, lower heteroalkyl, C1-4alkoxy, amino, cyano and halomethyl, wherein the #-N(R4)-R13-Z2b- substituent of formula (IIb) is attached to Ar2 at any Ar2 atom capable of being substituted;
Z1 is selected from N, CH, C-halo and C-CN;
Z2a, Z2b, and Z2c are each, independent from one another, selected from a bond, NR6, CR6aR6b, O, S, S(O), SO2, NR6C(O), NR6aC(O)NR6b, and NR6C(O)O;
R1 is selected from hydrogen, methyl, halo, halomethyl, ethyl and cyano;
R2 is selected from hydrogen, methyl, halo, halomethyl and cyano;
R3 is selected from hydrogen, lower alkyl and lower heteroalkyl;
R4 is selected from hydrogen, lower alkyl, monocyclic cycloalkyl, monocyclic heterocyclyl, and lower heteroalkyl or is taken together with an atom of R13 to form a cycloalkyl or heterocyclyl ring having between 3 and 7 ring atoms, wherein the lower alkyl, monocyclic cycloalkyl, monocyclic heterocyclyl, and lower heteroalkyl are optionally substituted with one or more halo, cyano, hydroxy, C1-4alkoxy, monocyclic cycloalkyl, monocyclic heterocyclyl, C(O)NR6aR6b, S(O)2NR6aR6b,
NHC(O)CHR6aR6b, NHS(O)CHR6aR6b, NHS(O)2CHR6aR6b, S(O)2CHR6aR6b or S(O)2NH2 groups;
R6, R6a and R6b are each, independent from one another, selected from hydrogen, lower alkyl, lower heteroalkyl, optionally substituted monocyclic cycloalkyl and monocyclic heterocyclyl, or are taken together with an atom from R13 to form a cycloalkyl or heterocyclyl ring having between 3 and 7 ring atoms;
R10 is selected from cyano, OR14, SR14, SOR14, SO2R14, SO2NR14aR14b, NR14aR14b, NHC(O)R14 and NHSO2R14; R11a and R11b are each, independently of one another, selected from hydrogen, halo, methyl, ethyl, halomethyl, hydroxyl, methoxy, CN, and SCH3;
R12 is selected from hydrogen, halo, cyano, lower alkyl, lower heteroalkyl, cycloalkyl, and heterocyclyl, wherein the alkyl, heteroalkyl, cycloalkyl, and heterocyclyl are optionally substituted with one or more halo, cyano, C1-4alkoxy, monocyclic cycloalkyl, monocyclic
heterocyclyl, NHC(O)CHR6aR6b, NHS(O)CHR6aR6b, NHS(O)2CHR6aR6b or S(O)2CHR6aR6b groups;
R13 is selected from a bond, optionally substituted lower alkylene, optionally substituted lower heteroalkylene, optionally substituted cycloalkyl or optionally substituted heterocyclyl;
R14 is selected from hydrogen, optionally substituted lower alkyl and optionally substituted lower heteroalkyl;
R14a and R14b are each, independently of one another, selected from hydrogen, optionally substituted lower alkyl, and optionally substituted lower heteroalkyl, or are taken together with the nitrogen atom to which they are bonded to form an optionally substituted monocyclic cycloalkyl or monocyclic heterocyclyl ring;
R15 is selected from hydrogen, halo, C1-6 alkanyl, C2-4 alkenyl, C2-4 alkynyl, and C1-4 haloalkyl and C1-4 hydroxyalkyl, with the proviso that when R15 is present, R4 is not C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C1-4 haloalkyl or C1-4 hydroxyalkyl, wherein the R4 C1-6 alkanyl, C2-4 alkenyl, C2- 4 alkynyl, C1-4 haloalkyl and C1-4 hydroxyalkyl are optionally substituted with one or more substituents independently selected from OCH3, OCH2CH2OCH3, and OCH2CH2NHCH3; and
# represents a point of attachment to a linker..
50. The ADC of claim 49, which is a compound according to structural formula (I):
Figure imgf000551_0001
wherein:
D is the Bcl-xL inhibitor drug of formula (IIa) or (IIb);
L is the linker;
Ab is the anti-hB7-H3 antibody;
LK represents a covalent linkage linking the linker (L) to the anti-hB7-H3 antibody (Ab); and
m is an integer ranging from 1 to 20.
51. The ADC of claim 49 or 50, in which Ar1 is unsubstituted.
52. The ADC of claim 51, in which Ar1 is
Figure imgf000552_0002
53. The ADC of claim 49 or 50, in which Ar2 is unsubstituted.
54. The ADC of claim 53, in which Ar2 is
Figure imgf000552_0003
which is optionally substituted at the 5-position with a group selected from hydroxyl, C1-4 alkoxy, and cyano; or
Figure imgf000552_0004
55. The ADC of claim 49 or 50, in which Z1 is N.
56. The ADC of claim 49 or 50, in which Z2a is O.
57. The ADC of claim 49 or 50, in which R1 is methyl or chloro.
58. The ADC of claim 49 or 50, in which R2 is hydrogen or methyl.
59. The ADC of claim 58, in which R2 is hydrogen.
60. The ADC of claim 49 or 50, in which R4 is hydrogen or lower alkyl, wherein the lower alkyl is optionally substituted with C1-4 alkoxy or C(O)NR6aR6b.
61. The ADC of claim 53, in which
Figure imgf000552_0001
substituted at the 5-position with a group selected from hydroxyl, C1-4 alkoxy, and cyano.
62. The ADC of claim 61, in which the drug is a Bcl-xL inhibitor according to structural formula (IIa).
63. The ADC of claim 49 or 50, in which the drug is a Bcl-xL inhibitor according to structural formula (IIa).
64. The ADC of claim 63, in which Z2a is CH2 or O.
65. The ADC of claim 63, in which R13 is selected from lower alkylene or lower heteroalkylene.
66. The ADC of claim 63, in which the group
Figure imgf000553_0001
is
Figure imgf000553_0002
.
67. The ADC of claim 63, in which the group
Figure imgf000553_0003
or
Figure imgf000553_0006
.
Figure imgf000553_0004
69. The ADC of claim 63, in which the group
Figure imgf000553_0005
.
70. The ADC of claim 62, wherein Z2a oxygen, R13 is CH2CH2, R4 is hydrogen or lower alkyl optionally substituted with C1-4 alkoxy or C(O)NR6aR6b.
71. The ADC of claim 52, which is a compound according to structural formula (IIb).
72. The ADC of claim 71, in which Z2b is a bond, O, or NR6, or and R13 is ethylene or optionally substituted heterocyclyl.
73. The ADC of claim 72, in which Z2c is O and R12 is lower alkyl optionally substituted with one or more halo or C1-4 alkoxy.
74. The ADC of claim 50, wherein the Bcl-xL inhibitor is selected from the group consisting of the following compounds modified in that the hydrogen corresponding to the # position of structural formula (IIa) or (IIb) is not present forming a monoradical:
6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4-tetrahydroquinolin-7-yl]-3-[1-({3,5-dimethyl- 7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid;
6-[4-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydro-2H-1,4-benzoxazin-6-yl]-3-[1-({3,5- dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4- yl]pyridine-2-carboxylic acid;
6-[4-(1,3-benzothiazol-2-ylcarbamoyl)-1-methyl-1,2,3,4-tetrahydroquinoxalin-6-yl]-3-[1- ({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol- 4-yl]pyridine-2-carboxylic acid;
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-5,6-dihydroimidazo[1,5-a]pyrazin-7(8H)- yl]pyridine-2-carboxylic acid;
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-hydroxy-3,4-dihydroisoquinolin-2(1H)- yl]pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid;
3-[1-({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl- 1H-pyrazol-4-yl]-6-[8-([1,3]thiazolo[5,4-b]pyridin-2-ylcarbamoyl)naphthalen-2-yl]pyridine-2- carboxylic acid;
3-[1-({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl- 1H-pyrazol-4-yl]-6-[8-([1,3]thiazolo[4,5-b]pyridin-2-ylcarbamoyl)naphthalen-2-yl]pyridine-2- carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-3-[1- ({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol- 4-yl]pyridine-2-carboxylic acid;
6-[5-(1,3-benzothiazol-2-ylcarbamoyl)quinolin-3-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid;
6-[4-(1,3-benzothiazol-2-ylcarbamoyl)quinolin-6-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid; 6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-3-{1- [(3-{2-[(2-methoxyethyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl- 1H-pyrazol-4-yl}pyridine-2-carboxylic acid;
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-cyano-3,4-dihydroisoquinolin-2(1H)- yl]pyridine-2-carboxylic acid;
6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-1,2,3,4-tetrahydroquinolin-7-yl]-3-{1-[(3-{2-[(2- methoxyethyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol- 4-yl}pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-3-{1-[(3-{2-[(2- methoxyethyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol- 4-yl}pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-[1-({3,5- dimethyl-7-[2-(oxetan-3-ylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4- yl]pyridine-2-carboxylic acid;
6-[6-(3-aminopyrrolidin-1-yl)-8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin- 2(1H)-yl]-3-(1-{[3-(2-methoxyethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-{1-[(3,5- dimethyl-7-{2-[(2-sulfamoylethyl)amino]ethoxy}tricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H- pyrazol-4-yl}pyridine-2-carboxylic acid;
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-[3-(1,3-benzothiazol-2-ylcarbamoyl)-6,7-dihydrothieno[3,2-c]pyridin-5(4H)- yl]pyridine-2-carboxylic acid;
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-3-(trifluoromethyl)-5,6-dihydroimidazo[1,5- a]pyrazin-7(8H)-yl]pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-6-{methyl[2-(methylamino)ethyl]amino}-3,4- dihydroisoquinolin-2(1H)-yl]-3-(1-{[3-(2-methoxyethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-6-methoxy-3,4-dihydroisoquinolin-2(1H)-yl]-3-[1- ({3,5-dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol- 4-yl]pyridine-2-carboxylic acid;
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-[4-(1,3-benzothiazol-2-ylcarbamoyl)quinolin-6-yl]pyridine-2-carboxylic acid; 6-[5-amino-8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-[1-({3,5- dimethyl-7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4- yl]pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-6-[3-(methylamino)prop-1-yn-1-yl]-3,4- dihydroisoquinolin-2(1H)-yl]-3-(1-{[3-(2-methoxyethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl]methyl}-5-methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid;
6-[4-(1,3-benzothiazol-2-ylcarbamoyl)isoquinolin-6-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid;
6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-1H-indol-2-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid;
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-1H-indol-2-yl]pyridine-2-carboxylic acid;
6-[7-(1,3-benzothiazol-2-ylcarbamoyl)-3-methyl-1H-indol-2-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-(1-{[3,5- dimethyl-7-(2-{[1-(methylsulfonyl)piperidin-4-yl]amino}ethoxy)tricyclo[3.3.1.13,7]dec-1-yl]methyl}- 5-methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-(1-{[3,5- dimethyl-7-(2-{[1-(methylsulfonyl)azetidin-3-yl]amino}ethoxy)tricyclo[3.3.1.13,7]dec-1-yl]methyl}- 5-methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid;
3-{1-[(3-{2-[(3-amino-3-oxopropyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl)methyl]-5-methyl-1H-pyrazol-4-yl}-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4- dihydroisoquinolin-2(1H)-yl]pyridine-2-carboxylic acid;
6-[3-(1,3-benzothiazol-2-ylcarbamoyl)-1H-indazol-5-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid;
6-[3-(1,3-benzothiazol-2-ylcarbamoyl)-1H-indol-5-yl]-3-[1-({3,5-dimethyl-7-[2- (methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid;
6-[3-(1,3-benzothiazol-2-ylcarbamoyl)-1H-pyrrolo[2,3-b]pyridin-5-yl]-3-[1-({3,5-dimethyl-7- [2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid; 6-(8-(benzo[d]thiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl)-3-(1-((3-(2-((2-(N,N- dimethylsulfamoyl)ethyl)amino)ethoxy)-5,7-dimethyladamantan-1-yl)methyl)-5-methyl-1H-pyrazol- 4-yl)picolinic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-3-{1-[(3-{2-[(3- hydroxypropyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol- 4-yl}pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-(1-{[3-(2-{[3- (dimethylamino)-3-oxopropyl]amino}ethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5- methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]-3-(1-{[3,5- dimethyl-7-(2-{[3-(methylamino)-3-oxopropyl]amino}ethoxy)tricyclo[3.3.1.13,7]dec-1-yl]methyl}-5- methyl-1H-pyrazol-4-yl)pyridine-2-carboxylic acid;
3-(1-{[3-(2-aminoacetamido)-5,7-dimethyltricyclo[3.3.1.13,7]decan-1-yl]methyl}-5-methyl- 1H-pyrazol-4-yl)-6-{8-[(1,3-benzothiazol-2-yl)carbamoyl]-3,4-dihydroisoquinolin-2(1H)-yl}pyridine- 2-carboxylic acid;
3-[1-({3-[(2-aminoethyl)sulfanyl]-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl- 1H-pyrazol-4-yl]-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]pyridine-2- carboxylic acid;
3-(1-{[3-(3-aminopropyl)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]pyridine-2- carboxylic acid; and
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]decan-1-yl]methyl}-5-methyl-1H- pyrazol-4-yl)-6-{5-[(1,3-benzothiazol-2-yl)carbamoyl]quinolin-3-yl}pyridine-2-carboxylic acid.
75. The ADC of any one of claims 49-74, in which the linker is cleavable by a lysosomal enzyme.
76. The ADC of claim 75, in which the lysosomal enzyme is Cathepsin B.
77. The ADC of anyone of claims 49-74, in which the linker comprises a segment according to structural formula (IVa), (IVb), (IVc), or (IVd):
Figure imgf000557_0001
Figure imgf000558_0001
Figure imgf000558_0002
Figure imgf000558_0003
wherein:
peptide represents a peptide (illustrated N→C, wherein peptide includes the amino and carboxy“termini”) a cleavable by a lysosomal enzyme;
T represents a polymer comprising one or more ethylene glycol units or an alkylene chain, or combinations thereof;
Ra is selected from hydrogen, C1-6 alkyl, SO3H and CH2SO3H;
Ry is hydrogen or C1-4 alkyl-(O)r-(C1-4 alkylene)s-G1 or C1-4 alkyl-(N)-[(C1-4 alkylene)- G1]2;
Rz is C1-4 alkyl-(O)r-(C1-4 alkylene)s-G2;
G1 is SO3H, CO2H, PEG 4-32, or sugar moiety;
G2 is SO3H, CO2H, or PEG 4-32 moiety;
r is 0 or 1;
s is 0 or 1;
p is an integer ranging from 0 to 5;
q is 0 or 1;
x is 0 or 1;
y is 0 or 1;
represents the point of attachment of the linker to the Bcl-xL inhibitor; and
* represents the point of attachment to the remainder of the linker.
78. The ADC of claim 77, in which peptide is selected from the group consisting of Val- Cit; Cit-Val; Ala-Ala; Ala-Cit; Cit-Ala; Asn-Cit; Cit-Asn; Cit-Cit; Val-Glu; Glu-Val; Ser-Cit; Cit-Ser; Lys-Cit; Cit-Lys; Asp-Cit; Cit-Asp; Ala-Val; Val-Ala; Phe-Lys; Lys-Phe; Val-Lys; Lys-Val; Ala-Lys; Lys-Ala; Phe-Cit; Cit-Phe; Leu-Cit; Cit-Leu; Ile-Cit; Cit-Ile; Phe-Arg; Arg-Phe; Cit-Trp; and Trp-Cit.
79. The ADC of claim 75, in which the lysosomal enzyme is β-glucuronidase or β-galactosidase.
80. The ADC of anyone of claims 49-74, in which the linker comprises a segment according to structural formula (Va), (Vb), (Vc), (Vd), or (Ve):
Figure imgf000559_0001
Figure imgf000559_0002
Figure imgf000559_0003
Figure imgf000560_0001
Figure imgf000560_0002
wherein:
q is 0 or 1;
r is 0 or 1;
X1 is CH2, O or NH;
represents the point of attachment of the linker to the drug; and
* represents the point of attachment to the remainder of the linker.
81. The ADC of anyone of claims 49-74, in which the linker comprises a segment according to structural formula (VIIIa), (VIIIb), or (VIIIc):
(VIIIa)
Figure imgf000560_0003
(VIIIb)
(VIIIc)
Figure imgf000561_0001
or a hydrolyzed derivative thereof, wherein:
Rq is H or–O-(CH2CH2O)11-CH3;
x is 0 or 1;
y is 0 or 1;
G3 is–CH2CH2CH2SO3H or–CH2CH2O-(CH2CH2O)11-CH3;
Rw is–O-CH2CH2SO3H or–NH(CO)-CH2CH2O-(CH2CH2O)12-CH3;
* represents the point of attachment to the remainder of the linker; and
represents the point of attachment of the linker to the antibody, wherein when in the hydrolyzed form, can be either at the α-position or β-position of the carboxylic acid next to it.
82. The ADC of anyone of claims 49-74, in which the linker comprises a polyethylene glycol segment having from 1 to 6 ethylene glycol units.
83. The ADC of anyone of claims 50-74, in which m is 2, 3 or 4.
84. The ADC of claim 83, in which linker L comprises a segment according to structural formula (IVa) or (IVb).
85. The ADC of anyone of claims 49-74, in which linker L is selected from the group consisting of IVa.1-IVa.8, IVb.1-IVb.19, IVc.1-IVc.7, IVd.1-IVd.4, Va.1-Va.12, Vb.1-Vb.10, Vc.1- Vc.11, Vd.1-Vd.6, Ve.1-Ve.2, VIa.1, VIc.1-V1c.2, VId.1-VId.4, VIIa.1-VIIa.4, VIIb.1-VIIb.8, VIIc.1-VIIc.6 in either the closed or open form.
86. The ADC of anyone of claims 49-74, in which the linker L is selected from the group consisting of IVb.2, IVc.5, IVc.6, IVc.7, IVd.4, Vb.9, Vc.11, VIIa.1, VIIa.3, VIIc.1, VIIc.4, and VIIc.5, wherein the maleimide of each linker has reacted with the antibody Ab, forming a covalent attachment as either a succinimide (closed form) or succinamide (open form).
87. The ADC of anyone of claims 49-74, in which the linker L is selected from the group consisting of IVb.2, IVc.5, IVc.6, IVd.4, Vc.11, VIIa.1, VIIa.3, VIIc.1, VIIc.4, VIIc.5, wherein the maleimide of each linker has reacted with the antibody Ab, forming a covalent attachment as either a succinimide (closed form) or succinamide (open form).
88. The ADC of anyone of claims 49-74, in which the linker L is selected from the group consisting of IVb.2, Vc.11, VIIa.3, IVc.6, and VIIc.1, wherein is the attachment point to drug D and @ is the attachment point to the LK, wherein when the linker is in the open form as shown below,
,
,
Figure imgf000562_0001
,
,
Figure imgf000563_0001
Figure imgf000564_0001
.
89. The ADC of anyone of claims 50-74, in which LK is a linkage formed with an amino group on the anti-hB7-H3 antibody Ab.
90. The ADC of claim 88, in which LK is an amide or a thiourea.
91. The ADC of anyone of claims 50-74, in which LK is a linkage formed with a sulfhydryl group on the anti-hB7-H3 antibody Ab.
92. The ADC of claim 91, in which LK is a thioether.
93. The ADC of anyone of claims 50-74, in which: LK is selected from the group consisting of amide, thiourea and thioether; and
m is an integer ranging from 1 to 8.
94. The ADC of claim 50, in which:
D is the Bcl-xL inhibitor as defined in claim 74;
L is selected from the group consisting of linkers IVa.1-IVa.8, IVb.1-IVb.19, IVc.1-IVc.7, IVd.1-IVd.4, Va.1-Va.12, Vb.1-Vb.10, Vc.1-Vc.11, Vd.1-Vd.6, Ve.1-Ve.2, VIa.1, VIc.1-V1c.2, VId.1-VId.4, VIIa.1-VIIa.4, VIIb.1-VIIb.8, and VIIc.1-VIIc.6, wherein each linker has reacted with the antibody, Ab, forming a covalent attachment;
LK is thioether; and
m is an integer ranging from 1 to 8.
95. The ADC of claim 50, in which:
D is the Bcl-xL inhibitor selected from the group consisting of the following compounds modified in that the hydrogen corresponding to the # position of structural formula (IIa) or (IIb) is not present, forming a monoradical:
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5- methyl-1H-pyrazol-4-yl)-6-[1-(1,3-benzothiazol-2-ylcarbamoyl)-5,6-dihydroimidazo[1,5-a]pyrazin- 7(8H)-yl]pyridine-2-carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)naphthalen-2-yl]-3-[1-({3,5-dimethyl- 7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid;
6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-methoxy-3,4-dihydroisoquinolin- 2(1H)-yl]-3-{1-[(3-{2-[(2-methoxyethyl)amino]ethoxy}-5,7-dimethyltricyclo[3.3.1.13,7]dec-1- yl)methyl]-5-methyl-1H-pyrazol-4-yl}pyridine-2-carboxylic acid;
3-(1-{[3-(2-aminoethoxy)-5,7-dimethyltricyclo[3.3.1.13,7]dec-1-yl]methyl}-5- methyl-1H-pyrazol-4-yl)-6-[8-(1,3-benzothiazol-2-ylcarbamoyl)-5-cyano-3,4-dihydroisoquinolin- 2(1H)-yl]pyridine-2-carboxylic acid;
6-[4-(1,3-benzothiazol-2-ylcarbamoyl)isoquinolin-6-yl]-3-[1-({3,5-dimethyl- 7-[2-(methylamino)ethoxy]tricyclo[3.3.1.13,7]dec-1-yl}methyl)-5-methyl-1H-pyrazol-4-yl]pyridine-2- carboxylic acid; and
3-{1-[(3-{2-[(3-amino-3-oxopropyl)amino]ethoxy}-5,7- dimethyltricyclo[3.3.1.13,7]dec-1-yl)methyl]-5-methyl-1H-pyrazol-4-yl}-6-[8-(1,3-benzothiazol-2- ylcarbamoyl)-3,4-dihydroisoquinolin-2(1H)-yl]pyridine-2-carboxylic acid;
L is selected from the group consisting of linkers IVb.2, IVc.5, IVc.6, IVc.7, IVd.4, Vb.9, Vc.11, VIIa.1, VIIa.3, VIIc.1, VIIc.4, and VIIc.5 in either closed or open forms;
LK is thioether; and
m is an integer ranging from 2 to 4
96. The ADC of claim 50, selected from the group consisting of formulae i-viii:
Figure imgf000566_0001
(iii),
Figure imgf000567_0001
Figure imgf000568_0001
,
(vii),
Figure imgf000569_0001
Figure imgf000570_0001
,
Figure imgf000571_0001
Figure imgf000572_0001
wherein m is an integer from 1 to 6.
97. The ADC of claim 96, wherein m is an integer from 2 to 6.
98. The ADC of any one of claims 49-97, wherein the anti-hB7-H3 antibody comprises a heavy chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 12, a heavy chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 140, and a heavy chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 10; a light chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 15, a light chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 7, and a light chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 136 or 138.
99. The ADC of any one of claims 49-97, wherein the antibody comprises a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 139, and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 135.
100. The ADC of any one of claims 49-97, wherein the antibody comprises a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 139, and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 137
101. The ADC of any one of claims 49-97, wherein the antibody comprises a light chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 39, a light chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 38, and a light chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 37; and a heavy chain CDR3 domain comprising the amino acid sequence set forth in SEQ ID NO: 35, a heavy chain CDR2 domain comprising the amino acid sequence set forth in SEQ ID NO: 34, and a heavy chain CDR1 domain comprising the amino acid sequence set forth in SEQ ID NO: 33.
102. The ADC of any one of claims 49-97, wherein the antibody comprises a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 147, and a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO: 144.
103. The ADC of claim 50, selected from the group consisting of huAb3v2.5-ZT, huAb3v2.5-ZZ, huAb3v2.5-XW, huAb3v2.5-SE, huAb3v2.5-SR, huAb3v2.5-YG, huAb3v2.5-KZ, huAb3v2.6-ZT, huAb3v2.6-ZZ, huAb3v2.6-XW, huAb3v2.6-SE, huAb3v2.6-SR, huAb3v2.6-YG, huAb3v2.6-KZ, huAb13v1-ZT, huAb13v1-ZZ, huAb13v1-XW, huAb13v1-SE, huAb13v1-SR, huAb13v1-YG, and huAb13v1-KZ.
104. A pharmaceutical composition comprising an effective amount of an ADC according to any one of claims 46-103 or 125, and a pharmaceutically acceptable carrier.
105. A pharmaceutical composition comprising an ADC mixture comprising a plurality of the ADC of any one of claims 46-103 or 125, and a pharmaceutically acceptable carrier.
106. The pharmaceutical composition of claim 105, wherein the ADC mixture has an average drug to antibody ratio (DAR) of 1.5 to 4.
107. The pharmaceutical composition of claim 105, wherein the ADC mixture comprises ADCs each having a DAR of 1.5 to 8.
108. A method for treating cancer, comprising administering a therapeutically effective amount of the ADC of any one of claims 46-103 or 125 to a subject in need thereof.
109. The method of claim 108, wherein the cancer is selected from the group consisting of small cell lung cancer, non-small cell lung cancer, breast cancer, ovarian cancer, a glioblastoma, prostate cancer, pancreatic cancer, colon cancer, gastric cancer, melanoma, hepatocellular carcinoma, head and neck cancer, acute myeloid leukemia (AML), non-Hodgkin's lymphoma (NHL), and kidney cancer.
110. The method of claim 108, wherein the cancer is a squamous cell carcinoma.
111. The method of claim 110, wherein the squamous cell carcinoma is squamous lung cancer or squamous head and neck cancer.
112. The method of claim 108, wherein the cancer is triple negative breast cancer.
113. The method of claim 108, wherein the cancer is non-small cell lung cancer.
114. A method for inhibiting or decreasing solid tumor growth in a subject having a solid tumor said method comprising administering an effective amount of the ADC of any one of claims 46-103 to the subject having the solid tumor, such that the solid tumor growth is inhibited or decreased.
115. The method of claim 114, wherein the solid tumor is a non-small cell lung carcinoma.
116. The method of any one of claims 109-115, wherein the ADC is administered in combination with an additional agent or an additional therapy.
117. The method of claim 116, wherein the additional agent is selected from the group consisting of an anti-PD1 antibody (e.g. pembrolizumab), an anti-PD-L1 antibody (e.g. atezolizumab), an anti-CTLA-4 antibody (e.g. ipilimumab), a MEK inhibitor (e.g. trametinib), an ERK inhibitor, a BRAF inhibitor (e.g. dabrafenib), osimertinib, erlotinib, gefitinib, sorafenib, a CDK9 inhibitor (e.g. dinaciclib), a MCL-1 inhibitor, temozolomide, a Bcl-xL inhibitor, Bcl-2 inhibitor (e.g. venetoclax), ibrutinib, a mTOR inhibitor (e.g. everolimus), a PI3K inhibitor (e.g. buparlisib), duvelisib, idelalisib, an AKT inhibitor, a HER2 inhibitor (e.g. lapatinib), a taxane (e.g. docetaxel, paclitaxel, nab- paclitaxel), an ADC comprising an auristatin, an ADC comprising a PBD (e.g. rovalpituzumab tesirine), an ADC comprising a maytansinoid (e.g. TDM1), a TRAIL agonist, a proteasome inhibitor (e.g. bortezomib), and a nicotinamide phosphoribosyltransferase (NAMPT) inhibitor.
118. The method of claim 116, wherein the additional therapy is radiation.
119. The method of claim 118, wherein the additional agent is a chemotherapeutic agent.
120. The method of any one of claims 108-113, wherein the cancer is characterized as having an activating EGFR mutation.
121. The method of claim 120, wherein the activating EGFR mutation is selected from the group consisting of an exon 19 deletion mutation, a single-point substitution mutation L858R in exon 21, a T790M point mutation, and combinations thereof.
122. An antibody, or antigen-binding portion thereof, that binds to hB7-H3 comprising a heavy chain variable region comprising a CDR set of SEQ ID NOs: 33, 35, and 35, and a light chain variable region comprising a CDR set of SEQ ID NOs: 37, 38, and 39.
123. The antibody, or antigen binding portion thereof, of claim 121, wherein the antibody, or antigen binding portion thereof, is humanized.
124. The antibody, or antigen binding portion thereof, according to claim 122, further comprising a human acceptor framework.
125. The antibody, or antigen binding portion thereof, of claim 123, wherein said human acceptor framework comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 156, 158, 166 and 167.
126. The ADC of any one of claims 49-97, wherein the antibody is an IgG having four polypeptide chains which are two heavy chains and two light chains.
127. A process for the preparation of an ADC according to structural formula (I):
Figure imgf000575_0001
wherein:
D is the Bcl-xL inhibitor drug of formula (IIa) or (IIb);
L is the linker;
Ab is the hB7-H3 antibody, wherein the hB7-H3 antibody comprises the heavy and light chain CDRs of huAb5v2.5, huAb5v2.6, of huAb13v1;
LK represents a covalent linkage linking linker L to antibody Ab; and
m is an integer ranging from 1 to 20.
the process comprising:
treating an antibody in an aqueous solution with an effective amount of a disulfide reducing agent at 30-40 °C for at least 15 minutes, and then cooling the antibody solution to 20-27 °C;
adding to the reduced antibody solution a solution of water/dimethyl sulfoxide comprising a synthon selected from the group of 2.1 to 2.31 and 2.34 to 2.72 (Table B);
adjusting the pH of the solution to a pH of 7.5 to 8.5; and
allowing the reaction to run for 48 to 80 hours to form the ADC;
wherein the mass is shifted by 18 ± 2 amu for each hydrolysis of a succinimide to a succinamide as measured by electron spray mass spectrometry; and
wherein the ADC is optionally purified by hydrophobic interaction chromatography.
128. The process of claim 127, wherein m is 2.
129. An ADC prepared by the process of claim 127 or 128.
PCT/US2017/036449 2016-06-08 2017-06-07 Anti-b7-h3 antibodies and antibody drug conjugates WO2017214339A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
BR112018075649-0A BR112018075649A2 (en) 2016-06-08 2017-06-07 anti-b7-h3 antibodies and antibody drug conjugates
CN201780048428.5A CN109641962A (en) 2016-06-08 2017-06-07 Anti- B7-H3 antibody and antibody drug conjugates
EP17732658.4A EP3469000A1 (en) 2016-06-08 2017-06-07 Anti-b7-h3 antibodies and antibody drug conjugates
AU2017279554A AU2017279554A1 (en) 2016-06-08 2017-06-07 Anti-B7-H3 antibodies and antibody drug conjugates
US16/308,742 US20200338209A1 (en) 2016-06-08 2017-06-07 Anti-b7-h3 antibodies and antibody drug conjugates
JP2018564199A JP2019521973A (en) 2016-06-08 2017-06-07 Anti-BH7-H3 antibody and antibody drug conjugate
CA3027103A CA3027103A1 (en) 2016-06-08 2017-06-07 Anti-b7-h3 antibodies and antibody drug conjugates
MX2018015285A MX2018015285A (en) 2016-06-08 2017-06-07 Anti-b7-h3 antibodies and antibody drug conjugates.

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201662347394P 2016-06-08 2016-06-08
US62/347,394 2016-06-08
US201662366478P 2016-07-25 2016-07-25
US62/366,478 2016-07-25

Publications (2)

Publication Number Publication Date
WO2017214339A1 true WO2017214339A1 (en) 2017-12-14
WO2017214339A4 WO2017214339A4 (en) 2018-02-08

Family

ID=59153290

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/036449 WO2017214339A1 (en) 2016-06-08 2017-06-07 Anti-b7-h3 antibodies and antibody drug conjugates

Country Status (9)

Country Link
US (1) US20200338209A1 (en)
EP (1) EP3469000A1 (en)
JP (1) JP2019521973A (en)
CN (1) CN109641962A (en)
AU (1) AU2017279554A1 (en)
BR (1) BR112018075649A2 (en)
CA (1) CA3027103A1 (en)
MX (1) MX2018015285A (en)
WO (1) WO2017214339A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020018964A1 (en) 2018-07-20 2020-01-23 Fred Hutchinson Cancer Research Center Compositions and methods for controlled expression of antigen-specific receptors
US10640563B2 (en) 2016-06-08 2020-05-05 Abbvie Inc. Anti-B7-H3 antibodies and antibody drug conjugates
WO2020236817A2 (en) 2019-05-20 2020-11-26 Novartis Ag Mcl-1 inhibitor antibody-drug conjugates and methods of use
CN113507941A (en) * 2018-12-21 2021-10-15 萨普雷米科技有限公司 Bioactive molecular cluster
JP2022501076A (en) * 2018-09-26 2022-01-06 フーヂョウ ティーセルテック バイオテクノロジー カンパニー リミテッドFuzhou Tcelltech Biotechnology Co., Ltd. Monoclonal antibody against B7-H3 and its use in cell therapy
WO2023160376A1 (en) * 2022-02-25 2023-08-31 Nanjing Probio Biotech Co., Ltd. Antibodies and variants thereof against human b7-h3
US11759527B2 (en) 2021-01-20 2023-09-19 Abbvie Inc. Anti-EGFR antibody-drug conjugates
JP7458997B2 (en) 2018-05-29 2024-04-01 ブリストル-マイヤーズ スクイブ カンパニー Modified self-cleaving moieties for use in prodrugs and conjugates and methods of use and preparation thereof

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117024591A (en) * 2020-04-22 2023-11-10 复星凯特生物科技有限公司 Monoclonal antibody against human B7-H3 and application thereof
WO2022104692A1 (en) * 2020-11-20 2022-05-27 Bliss Biopharmaceutical (Hangzhou) Co., Ltd. Engineered antibody, antibody-drug conjugate, and use thereof
WO2022194078A1 (en) * 2021-03-15 2022-09-22 北京大学 Conjugate for target molecule modification and preparation method therefor
WO2022232630A1 (en) * 2021-04-30 2022-11-03 Board Of Regents, The University Of Texas System Use of carboxylates for carbon sequestration, improved oil recovery, and hydrogen storage and reproduction
CN113274502B (en) * 2021-05-05 2023-01-03 中山大学肿瘤防治中心(中山大学附属肿瘤医院、中山大学肿瘤研究所) Compositions for specific type three-negative breast cancer immunotherapy
WO2023236949A1 (en) * 2022-06-07 2023-12-14 映恩生物制药(苏州)有限公司 Anti-b7h4 antibody-drug conjugate and use thereof

Citations (169)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4486414A (en) 1983-03-21 1984-12-04 Arizona Board Of Reagents Dolastatins A and B cell growth inhibitory substances
EP0077671B1 (en) 1981-10-19 1986-06-25 Ortho Diagnostic Systems Inc. Immunological reagents employing polymeric backbones
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
EP0229246A2 (en) 1986-01-15 1987-07-22 ANT Nachrichtentechnik GmbH Method for decoding digital signals, as well as a Viterbi decoder and applications
EP0239400A2 (en) 1986-03-27 1987-09-30 Medical Research Council Recombinant antibodies and methods for their production
US4814470A (en) 1986-07-17 1989-03-21 Rhone-Poulenc Sante Taxol derivatives, their preparation and pharmaceutical compositions containing them
US4816397A (en) 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
US4816444A (en) 1987-07-10 1989-03-28 Arizona Board Of Regents, Arizona State University Cell growth inhibitory substance
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4879278A (en) 1989-05-16 1989-11-07 Arizona Board Of Regents Isolation and structural elucidation of the cytostatic linear depsipeptide dolastatin 15
US4880935A (en) 1986-07-11 1989-11-14 Icrf (Patents) Limited Heterobifunctional linking agents derived from N-succinimido-dithio-alpha methyl-methylene-benzoates
WO1989012624A2 (en) 1988-06-14 1989-12-28 Cetus Corporation Coupling agents and sterically hindered disulfide linked conjugates prepared therefrom
WO1990005144A1 (en) 1988-11-11 1990-05-17 Medical Research Council Single domain ligands, receptors comprising said ligands, methods for their production, and use of said ligands and receptors
US4942184A (en) 1988-03-07 1990-07-17 The United States Of America As Represented By The Department Of Health And Human Services Water soluble, antineoplastic derivatives of taxol
US4960790A (en) 1989-03-09 1990-10-02 University Of Kansas Derivatives of taxol, pharmaceutical compositions thereof and methods for the preparation thereof
WO1990014443A1 (en) 1989-05-16 1990-11-29 Huse William D Co-expression of heteromeric receptors
WO1990014424A1 (en) 1989-05-16 1990-11-29 Scripps Clinic And Research Foundation Method for isolating receptors having a preselected specificity
WO1990014430A1 (en) 1989-05-16 1990-11-29 Scripps Clinic And Research Foundation A new method for tapping the immunological repertoire
US4978744A (en) 1989-01-27 1990-12-18 Arizona Board Of Regents Synthesis of dolastatin 10
US4986988A (en) 1989-05-18 1991-01-22 Arizona Board Of Regents Isolation and structural elucidation of the cytostatic linear depsipeptides dolastatin 13 and dehydrodolastatin 13
WO1991009967A1 (en) 1989-12-21 1991-07-11 Celltech Limited Humanised antibodies
US5076973A (en) 1988-10-24 1991-12-31 Arizona Board Of Regents Synthesis of dolastatin 3
US5122368A (en) 1988-02-11 1992-06-16 Bristol-Myers Squibb Company Anthracycline conjugates having a novel linker and methods for their production
US5124471A (en) 1990-03-26 1992-06-23 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Bifunctional dtpa-type ligand
US5138036A (en) 1989-11-13 1992-08-11 Arizona Board Of Regents Acting On Behalf Of Arizona State University Isolation and structural elucidation of the cytostatic cyclodepsipeptide dolastatin 14
US5157049A (en) 1988-03-07 1992-10-20 The United States Of America As Represented By The Department Of Health & Human Services Method of treating cancers sensitive to treatment with water soluble derivatives of taxol
WO1992022332A2 (en) 1991-06-17 1992-12-23 The Regents Of The University Of California Drug delivery of antisense oligonucleotides and peptides to tissues in vivo and to cells using avidin-biotin technology
EP0519596A1 (en) 1991-05-17 1992-12-23 Merck & Co. Inc. A method for reducing the immunogenicity of antibody variable domains
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
WO1993021232A1 (en) 1992-04-10 1993-10-28 Research Development Foundation IMMUNOTOXINS DIRECTED AGAINST c-erbB-2 (HER-2/neu) RELATED SURFACE ANTIGENS
US5278324A (en) 1990-08-28 1994-01-11 Virginia Tech Intellectual Properties, Inc. Water soluble derivatives of taxol
EP0592106A1 (en) 1992-09-09 1994-04-13 Immunogen Inc Resurfacing of rodent antibodies
WO1994016729A1 (en) 1993-01-28 1994-08-04 Neorx Corporation Targeted nitric oxide pathway or nitric oxide synthase modulation
US5362831A (en) 1992-06-19 1994-11-08 Farmitalia Carlo Erba S.R.L. Polymer-bound paclitaxel derivatives
US5380751A (en) 1992-12-04 1995-01-10 Bristol-Myers Squibb Company 6,7-modified paclitaxels
US5399363A (en) 1991-01-25 1995-03-21 Eastman Kodak Company Surface modified anticancer nanoparticles
US5403858A (en) 1991-07-08 1995-04-04 Rhone-Poulenc Rorer, S.A. New compositions containing taxane derivatives
US5407683A (en) 1990-06-01 1995-04-18 Research Corporation Technologies, Inc. Pharmaceutical solutions and emulsions containing taxol
US5410024A (en) 1993-01-21 1995-04-25 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide amides
US5415869A (en) 1993-11-12 1995-05-16 The Research Foundation Of State University Of New York Taxol formulation
US5424073A (en) 1992-03-23 1995-06-13 Georgetown University Liposome encapsulated taxol and a method of using the same
WO1995015770A1 (en) 1993-12-09 1995-06-15 Neorx Corporation Pretargeting methods and compounds
US5433364A (en) 1993-02-19 1995-07-18 Dynetics Engineering Corporation Card package production system with burster and carrier verification apparatus
US5438072A (en) 1992-12-02 1995-08-01 Rhone-Poulenc Rorer S.A. Taxoid-based compositions
US5504191A (en) 1994-08-01 1996-04-02 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide methyl esters
US5521284A (en) 1994-08-01 1996-05-28 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide amides and esters
US5530097A (en) 1994-08-01 1996-06-25 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory peptide amides
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5554725A (en) 1994-09-14 1996-09-10 Arizona Board Of Regents Acting On Behalf Of Arizona State University Synthesis of dolastatin 15
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5599902A (en) 1994-11-10 1997-02-04 Arizona Board Of Regents Acting On Behalf Of Arizona State University Cancer inhibitory peptides
US5622929A (en) 1992-01-23 1997-04-22 Bristol-Myers Squibb Company Thioether conjugates
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5635483A (en) 1992-12-03 1997-06-03 Arizona Board Of Regents Acting On Behalf Of Arizona State University Tumor inhibiting tetrapeptide bearing modified phenethyl amides
US5641803A (en) 1992-08-03 1997-06-24 Bristol-Myers Squibb Company Methods for administration of taxol
WO1997029131A1 (en) 1996-02-09 1997-08-14 Basf Aktiengesellschaft HUMAN ANTIBODIES THAT BIND HUMAN TNF$g(a)
US5663149A (en) 1994-12-13 1997-09-02 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide heterocyclic and halophenyl amides
WO1997031655A2 (en) 1996-02-27 1997-09-04 Fondazione Centro San Raffaele Del Monte Tabor Modified cytokines for therapeutic use
US5665671A (en) 1994-07-21 1997-09-09 Hydrogeo North America Compound to defend plants from vegetal parasites
US5705503A (en) 1995-05-25 1998-01-06 Goodall; Brian Leslie Addition polymers of polycycloolefins containing functional substituents
US5712374A (en) 1995-06-07 1998-01-27 American Cyanamid Company Method for the preparation of substantiallly monomeric calicheamicin derivative/carrier conjugates
US5714352A (en) 1996-03-20 1998-02-03 Xenotech Incorporated Directed switch-mediated DNA recombination
US5714350A (en) 1992-03-09 1998-02-03 Protein Design Labs, Inc. Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
US5723323A (en) 1985-03-30 1998-03-03 Kauffman; Stuart Alan Method of identifying a stochastically-generated peptide, polypeptide, or protein having ligand binding property and compositions thereof
US5728687A (en) 1992-11-10 1998-03-17 Rhone-Poulenc Rorer, S.A. Antitumour compositions containing taxane derivatives
US5739116A (en) 1994-06-03 1998-04-14 American Cyanamid Company Enediyne derivatives useful for the synthesis of conjugates of methyltrithio antitumor agents
WO1998022451A1 (en) 1996-11-19 1998-05-28 Daiichi Pharmaceutical Co., Ltd. Taxol derivatives
US5763192A (en) 1986-11-20 1998-06-09 Ixsys, Incorporated Process for obtaining DNA, RNA, peptides, polypeptides, or protein, by recombinant DNA technique
US5766886A (en) 1991-12-13 1998-06-16 Xoma Corporation Modified antibody variable domains
US5770701A (en) 1987-10-30 1998-06-23 American Cyanamid Company Process for preparing targeted forms of methyltrithio antitumor agents
US5770710A (en) 1987-10-30 1998-06-23 American Cyanamid Company Antitumor and antibacterial substituted disulfide derivatives prepared from compounds possessing a methlytrithio group
US5773464A (en) 1996-09-30 1998-06-30 Bristol-Myers Squibb Company C-10 epoxy taxanes
WO1998028288A1 (en) 1996-12-24 1998-07-02 Bristol-Myers Squibb Company 6-thio-substituted paclitaxels
US5780588A (en) 1993-01-26 1998-07-14 Arizona Board Of Regents Elucidation and synthesis of selected pentapeptides
WO1998035704A1 (en) 1997-02-13 1998-08-20 Bone Care International, Inc. Targeted therapeutic delivery of vitamin d compounds
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
US5821263A (en) 1996-08-26 1998-10-13 Bristol-Myers Squibb Company Sulfenamide taxane derivatives
US5824805A (en) 1995-12-22 1998-10-20 King; Dalton Branched hydrazone linkers
US5840929A (en) 1995-04-14 1998-11-24 Bristol-Myers Squibb Company C4 methoxy ether derivatives of paclitaxel
US5869680A (en) 1992-10-05 1999-02-09 Rhone-Poulenc Rorer, S.A. Process for preparing taxane derivatives
WO1999009021A1 (en) 1997-08-18 1999-02-25 Florida State University Process for selective derivatization of taxanes
WO1999014209A1 (en) 1997-09-17 1999-03-25 Kabushiki Kaisha Yakult Honsha New taxane derivatives
WO1999018113A1 (en) 1997-10-08 1999-04-15 Bio Research Corporation Of Yokohama Taxoid derivatives and process for producing the same
WO1999019500A1 (en) 1997-10-10 1999-04-22 The Government Of The United States Of America, Represented By The Secretary, Department Of Healt H And Human Services Complex of biotinylated viral vector and ligand for targeted gene delivery
WO1999054342A1 (en) 1998-04-20 1999-10-28 Pablo Umana Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
US6034065A (en) 1992-12-03 2000-03-07 Arizona Board Of Regents Elucidation and synthesis of antineoplastic tetrapeptide phenethylamides of dolastatin 10
US6090382A (en) 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
WO2001009785A1 (en) 1999-07-31 2001-02-08 Kyu Jin Park Study method and apparatus using digital audio and caption data
US6204023B1 (en) 1985-11-01 2001-03-20 Xoma Ltd. Modular assembly of antibody genes, antibodies prepared thereby and use
US6214345B1 (en) 1993-05-14 2001-04-10 Bristol-Myers Squibb Co. Lysosomal enzyme-cleavable antitumor drug conjugates
US6239104B1 (en) 1997-02-25 2001-05-29 Arizona Board Of Regents Isolation and structural elucidation of the cytostatic linear and cyclo-depsipeptides dolastatin 16, dolastatin 17, and dolastatin 18
US6323315B1 (en) 1999-09-10 2001-11-27 Basf Aktiengesellschaft Dolastatin peptides
WO2001090198A1 (en) 2000-05-24 2001-11-29 Ludwig Institute For Cancer Research Multicomponent conjugates which bind to target molecules and stimulate cell lysis
EP1176195A1 (en) 1999-04-09 2002-01-30 Kyowa Hakko Kogyo Co., Ltd. Method for controlling the activity of immunologically functional molecule
US6410319B1 (en) 1998-10-20 2002-06-25 City Of Hope CD20-specific redirected T cells and their use in cellular immunotherapy of CD20+ malignancies
US6441163B1 (en) 2001-05-31 2002-08-27 Immunogen, Inc. Methods for preparation of cytotoxic conjugates of maytansinoids and cell binding agents
US20020137134A1 (en) 2000-06-28 2002-09-26 Gerngross Tillman U. Methods for producing modified glycoproteins
WO2002088172A2 (en) 2001-04-30 2002-11-07 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
WO2003016466A2 (en) 2001-08-17 2003-02-27 Eli Lilly And Company ANTI-Aβ ANTIBODIES
WO2003035835A2 (en) 2001-10-25 2003-05-01 Genentech, Inc. Glycoprotein compositions
US20030083263A1 (en) 2001-04-30 2003-05-01 Svetlana Doronina Pentapeptide compounds and uses related thereto
WO2003093793A2 (en) 2002-05-01 2003-11-13 Trellis Bioscience, Inc. Binary or polynary targeting and uses thereof
US20040018590A1 (en) 2000-06-28 2004-01-29 Gerngross Tillman U. Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
WO2004010957A2 (en) 2002-07-31 2004-02-05 Seattle Genetics, Inc. Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
WO2004050016A2 (en) 2002-11-27 2004-06-17 The Regents Of The University Of California Delivery of pharmaceutical agents via the human insulin receptor
US20050042664A1 (en) 2003-08-22 2005-02-24 Medimmune, Inc. Humanization of antibodies
US6913748B2 (en) 2002-08-16 2005-07-05 Immunogen, Inc. Cross-linkers with high reactivity and solubility and their use in the preparation of conjugates for targeted delivery of small molecule drugs
WO2005081898A2 (en) 2004-02-20 2005-09-09 The Trustees Of The University Of Pennsylvania Binding peptidomimetics and uses of the same
WO2005100584A2 (en) 2004-04-15 2005-10-27 Glycofi, Inc. Production of galactosylated glycoproteins in lower eukaryotes
US20050238649A1 (en) 2003-11-06 2005-10-27 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
US20050271615A1 (en) 2002-08-30 2005-12-08 Doron Shabat Self-immolative dendrimers releasing many active moieties upon a single activating event
US20060024317A1 (en) 2004-05-19 2006-02-02 Medarex, Inc Chemical linkers and conjugates thereof
US20060116422A1 (en) 2002-11-14 2006-06-01 De Groot Franciscus Marinus H Prodrugs built as multiple self-elimination-release spacers
WO2006083562A2 (en) 2005-02-02 2006-08-10 Awdalla, Essam, T. Method and multicomponent conjugates for treating cancer
WO2006089668A1 (en) 2005-02-23 2006-08-31 Ludwig-Maximilians-Universität Transport of nano- and macromolecular structures into cytoplasm and nucleus of cells
WO2006111759A1 (en) 2005-04-21 2006-10-26 Spirogen Limited Pyrrolobenzodiazepines
US7223837B2 (en) 2001-03-23 2007-05-29 Syntarga B.V. Elongated and multiple spacers in activatible prodrugs
US20070173497A1 (en) 2004-03-01 2007-07-26 Spirogen Limited 11-Hydroxy-5h-pyrrolo[2,1-c][1,4] benzodiazepin-5-one derivatives as key intermediates for the preparation of c2 substituted pyrrolobenzodiazepines
WO2007150020A1 (en) 2006-06-23 2007-12-27 Simon Paul M Targeted immune conjugates
WO2008097870A2 (en) 2007-02-02 2008-08-14 Baylor Research Institute Agents that engage antigen - presenting cells through dendritic cell asialoglycoprotein receptor (dc- asgpr)
WO2008097866A2 (en) 2007-02-02 2008-08-14 Baylor Research Institute Vaccines based on targeting antigen to dcir expressed an antigen-presenting cells
WO2008100624A2 (en) 2007-02-16 2008-08-21 Merrimack Pharmaceuticals, Inc. Antibodies against erbb3 and uses thereof
WO2008103947A2 (en) 2007-02-23 2008-08-28 Baylor Research Institute Activation of human antigen-presenting cells through clec-6
WO2008103953A2 (en) 2007-02-23 2008-08-28 Baylor Research Institute Activation of human antigen-presenting cells through dendritic cell lectin-like oxidized ldl receptor-1 (lox-1)
WO2008135237A1 (en) 2007-05-03 2008-11-13 Medizinische Universität Innsbruck Complement factor h-derived short consensus repeat-antibody constructs
US7598028B2 (en) 2006-11-28 2009-10-06 The Regents Of The University Of Michigan Compositions and methods for detecting and treating prostate disorders
US20090318668A1 (en) 2006-02-02 2009-12-24 Patrick Henry Beusker Water-Soluble CC-1065 Analogs and Their Conjugates
US20100152725A1 (en) 2008-12-12 2010-06-17 Angiodynamics, Inc. Method and system for tissue treatment utilizing irreversible electroporation and thermal track coagulation
WO2010111198A1 (en) 2009-03-23 2010-09-30 Quark Pharmaceuticals, Inc. Compounds compositions and methods of treating cancer and fibrotic diseases
WO2011000370A1 (en) 2009-06-30 2011-01-06 Röhm Gmbh Drilling device
US20110076232A1 (en) 2009-09-29 2011-03-31 Ludwig Institute For Cancer Research Specific binding proteins and uses thereof
WO2011057216A1 (en) 2009-11-06 2011-05-12 The Pennsylvania State Research Foundation Bioconjugation of calcium phosphosilicate nanoparticles for selective targeting of cells in vivo
WO2011058321A1 (en) 2009-11-11 2011-05-19 King's College Hospital Nhs Foundation Trust Conjugate molecule
US7989434B2 (en) 2004-02-23 2011-08-02 Seattle Genetics, Inc. Heterocyclic self-immolative linkers and conjugates
WO2011109400A2 (en) * 2010-03-04 2011-09-09 Macrogenics,Inc. Antibodies reactive with b7-h3, immunologically active fragments thereof and uses thereof
WO2011130598A1 (en) 2010-04-15 2011-10-20 Spirogen Limited Pyrrolobenzodiazepines and conjugates thereof
WO2012027494A1 (en) 2010-08-24 2012-03-01 Regents Of The University Of Minnesota Bispecific targeting reagents
US8142784B2 (en) 2004-06-01 2012-03-27 Genentech, Inc. Antibody-drug conjugates and methods
US20120107332A1 (en) 2005-07-18 2012-05-03 Seattle Genetics, Inc. Beta-Glucuronide-Linker Drug Conjugates
US8236319B2 (en) 2008-04-30 2012-08-07 Immunogen, Inc. Cross-linkers and their uses
US8309093B2 (en) 2009-03-06 2012-11-13 Agensys, Inc. Antibody drug conjugates (ADC) that bind to 24P4C12 proteins
US8349308B2 (en) 2010-03-26 2013-01-08 Mersana Therapeutics, Inc. Modified polymers for delivery of polynucleotides, method of manufacture, and methods of use thereof
US20130028919A1 (en) 2010-04-15 2013-01-31 Spirogen Developments Sàrl Targeted pyrrolobenzodiazapine conjugates
US8389282B2 (en) 2007-03-30 2013-03-05 Memorial Sloan-Kettering Cancer Center Constitutive expression of costimulatory ligands on adoptively transferred T lymphocytes
US8399512B2 (en) 2007-11-28 2013-03-19 Mersana Therapeutics, Inc. Biocompatible biodegradable fumagillin analog conjugates
US20130189218A1 (en) 2011-12-23 2013-07-25 Mersana Therapeutics, Inc. Pharmaceutical formulations for fumagillin derivative-phf conjugates
US20130224228A1 (en) 2011-12-05 2013-08-29 Igenica, Inc. Antibody-Drug Conjugates and Related Compounds, Compositions, and Methods
US8524214B2 (en) 2009-05-28 2013-09-03 Mersana Therapeutics, Inc. Polyal drug conjugates comprising variable rate-releasing linkers
US8535678B2 (en) 2003-02-20 2013-09-17 Seattle Genetics, Inc. Anti-CD70 antibody-drug conjugates and their use for the treatment of cancer and immune disorders
US20130303509A1 (en) 2012-05-11 2013-11-14 Abbvie Inc. Nampt inhibitors
WO2013173337A2 (en) 2012-05-15 2013-11-21 Seattle Genetics, Inc. Self-stabilizing linker conjugates
US20130309256A1 (en) 2012-05-15 2013-11-21 Seattle Genetics, Inc. Self-stabilizing linker conjugates
US20140017265A1 (en) 2012-07-05 2014-01-16 Mersana Therapeutics, Inc. Terminally Modified Polymers and Conjugates Thereof
WO2014093640A1 (en) 2012-12-12 2014-06-19 Mersana Therapeutics,Inc. Hydroxy-polmer-drug-protein conjugates
WO2014093394A1 (en) 2012-12-10 2014-06-19 Mersana Therapeutics, Inc. Protein-polymer-drug conjugates
WO2014093379A1 (en) 2012-12-10 2014-06-19 Mersana Therapeutics, Inc. Auristatin compounds and conjugates thereof
US8822647B2 (en) 2008-08-26 2014-09-02 City Of Hope Method and compositions using a chimeric antigen receptor for enhanced anti-tumor effector functioning of T cells
US20140286968A1 (en) 2013-03-15 2014-09-25 Abbvie Inc. Antibody drug conjugate (adc) purification
US20140322275A1 (en) 2013-02-20 2014-10-30 Jennifer Brogdon TREATMENT OF CANCER USING HUMANIZED ANTI-EGFRvIII CHIMERIC ANTIGEN RECEPTOR
US20140328750A1 (en) * 2010-03-04 2014-11-06 Macrogenics, Inc. Antibodies Reactive with B7-H3 and Uses Thereof
US8906682B2 (en) 2010-12-09 2014-12-09 The Trustees Of The University Of Pennsylvania Methods for treatment of cancer
US9105939B2 (en) 2008-03-10 2015-08-11 Nissan Motor Co., Ltd. Battery with battery electrode and method of manufacturing same
US9109630B2 (en) 2013-01-15 2015-08-18 Jtekt Corporation Rolling bearing unit
US20160158377A1 (en) 2014-12-09 2016-06-09 Abbvie Inc. BCL-XL Inhibitory Compounds and Antibody Drug Conjugates Including the Same
US9401234B2 (en) 2013-03-22 2016-07-26 Polytronics Technology Corp. Over-current protection device
US9618978B2 (en) 2013-10-03 2017-04-11 Acer Incorporated Methods for controlling a touch panel and portable computers using the same
US9816280B1 (en) 2016-11-02 2017-11-14 Matthew Reitnauer Portable floor

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
LT2703486T (en) * 2011-04-25 2018-05-25 Daiichi Sankyo Company, Limited Anti-b7-h3 antibody

Patent Citations (192)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0077671B1 (en) 1981-10-19 1986-06-25 Ortho Diagnostic Systems Inc. Immunological reagents employing polymeric backbones
US4486414A (en) 1983-03-21 1984-12-04 Arizona Board Of Reagents Dolastatins A and B cell growth inhibitory substances
US4816397A (en) 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
US5817483A (en) 1985-03-30 1998-10-06 Stuart Kauffman Process for the production of stochastically-generated peptides,polypeptides or proteins having a predetermined property
US5814476A (en) 1985-03-30 1998-09-29 Stuart Kauffman Process for the production of stochastically-generated transcription or translation products
US5976862A (en) 1985-03-30 1999-11-02 Ixsys Corporation Process for obtaining DNA, RNA, peptides, polypeptides, or proteins, by recombinant DNA technique
US5723323A (en) 1985-03-30 1998-03-03 Kauffman; Stuart Alan Method of identifying a stochastically-generated peptide, polypeptide, or protein having ligand binding property and compositions thereof
US5824514A (en) 1985-03-30 1998-10-20 Stuart A. Kauffman Process for the production of expression vectors comprising at least one stochastic sequence of polynucleotides
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US6204023B1 (en) 1985-11-01 2001-03-20 Xoma Ltd. Modular assembly of antibody genes, antibodies prepared thereby and use
EP0229246A2 (en) 1986-01-15 1987-07-22 ANT Nachrichtentechnik GmbH Method for decoding digital signals, as well as a Viterbi decoder and applications
EP0239400A2 (en) 1986-03-27 1987-09-30 Medical Research Council Recombinant antibodies and methods for their production
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US4880935A (en) 1986-07-11 1989-11-14 Icrf (Patents) Limited Heterobifunctional linking agents derived from N-succinimido-dithio-alpha methyl-methylene-benzoates
US4814470A (en) 1986-07-17 1989-03-21 Rhone-Poulenc Sante Taxol derivatives, their preparation and pharmaceutical compositions containing them
US5763192A (en) 1986-11-20 1998-06-09 Ixsys, Incorporated Process for obtaining DNA, RNA, peptides, polypeptides, or protein, by recombinant DNA technique
US5648260A (en) 1987-03-18 1997-07-15 Scotgen Biopharmaceuticals Incorporated DNA encoding antibodies with altered effector functions
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US4816444A (en) 1987-07-10 1989-03-28 Arizona Board Of Regents, Arizona State University Cell growth inhibitory substance
US5770710A (en) 1987-10-30 1998-06-23 American Cyanamid Company Antitumor and antibacterial substituted disulfide derivatives prepared from compounds possessing a methlytrithio group
US5770701A (en) 1987-10-30 1998-06-23 American Cyanamid Company Process for preparing targeted forms of methyltrithio antitumor agents
US5122368A (en) 1988-02-11 1992-06-16 Bristol-Myers Squibb Company Anthracycline conjugates having a novel linker and methods for their production
US5157049A (en) 1988-03-07 1992-10-20 The United States Of America As Represented By The Department Of Health & Human Services Method of treating cancers sensitive to treatment with water soluble derivatives of taxol
US4942184A (en) 1988-03-07 1990-07-17 The United States Of America As Represented By The Department Of Health And Human Services Water soluble, antineoplastic derivatives of taxol
WO1989012624A2 (en) 1988-06-14 1989-12-28 Cetus Corporation Coupling agents and sterically hindered disulfide linked conjugates prepared therefrom
US5076973A (en) 1988-10-24 1991-12-31 Arizona Board Of Regents Synthesis of dolastatin 3
WO1990005144A1 (en) 1988-11-11 1990-05-17 Medical Research Council Single domain ligands, receptors comprising said ligands, methods for their production, and use of said ligands and receptors
US5693762A (en) 1988-12-28 1997-12-02 Protein Design Labs, Inc. Humanized immunoglobulins
US6180370B1 (en) 1988-12-28 2001-01-30 Protein Design Labs, Inc. Humanized immunoglobulins and methods of making the same
US5585089A (en) 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US4978744A (en) 1989-01-27 1990-12-18 Arizona Board Of Regents Synthesis of dolastatin 10
US4960790A (en) 1989-03-09 1990-10-02 University Of Kansas Derivatives of taxol, pharmaceutical compositions thereof and methods for the preparation thereof
US4879278A (en) 1989-05-16 1989-11-07 Arizona Board Of Regents Isolation and structural elucidation of the cytostatic linear depsipeptide dolastatin 15
WO1990014430A1 (en) 1989-05-16 1990-11-29 Scripps Clinic And Research Foundation A new method for tapping the immunological repertoire
WO1990014424A1 (en) 1989-05-16 1990-11-29 Scripps Clinic And Research Foundation Method for isolating receptors having a preselected specificity
WO1990014443A1 (en) 1989-05-16 1990-11-29 Huse William D Co-expression of heteromeric receptors
US4986988A (en) 1989-05-18 1991-01-22 Arizona Board Of Regents Isolation and structural elucidation of the cytostatic linear depsipeptides dolastatin 13 and dehydrodolastatin 13
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US5138036A (en) 1989-11-13 1992-08-11 Arizona Board Of Regents Acting On Behalf Of Arizona State University Isolation and structural elucidation of the cytostatic cyclodepsipeptide dolastatin 14
WO1991009967A1 (en) 1989-12-21 1991-07-11 Celltech Limited Humanised antibodies
US5434287A (en) 1990-03-26 1995-07-18 The United States Of America As Represented By The Department Of Health And Human Services Bifunctional DTPA-type ligand
US5124471A (en) 1990-03-26 1992-06-23 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Bifunctional dtpa-type ligand
US5286850A (en) 1990-03-26 1994-02-15 The United States Of America As Represented By The Department Of Health And Human Services Antibody DTPA-type ligand conjugates
US5407683A (en) 1990-06-01 1995-04-18 Research Corporation Technologies, Inc. Pharmaceutical solutions and emulsions containing taxol
US5278324A (en) 1990-08-28 1994-01-11 Virginia Tech Intellectual Properties, Inc. Water soluble derivatives of taxol
US5399363A (en) 1991-01-25 1995-03-21 Eastman Kodak Company Surface modified anticancer nanoparticles
EP0519596A1 (en) 1991-05-17 1992-12-23 Merck & Co. Inc. A method for reducing the immunogenicity of antibody variable domains
WO1992022332A2 (en) 1991-06-17 1992-12-23 The Regents Of The University Of California Drug delivery of antisense oligonucleotides and peptides to tissues in vivo and to cells using avidin-biotin technology
US5403858A (en) 1991-07-08 1995-04-04 Rhone-Poulenc Rorer, S.A. New compositions containing taxane derivatives
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5766886A (en) 1991-12-13 1998-06-16 Xoma Corporation Modified antibody variable domains
US5622929A (en) 1992-01-23 1997-04-22 Bristol-Myers Squibb Company Thioether conjugates
US6350861B1 (en) 1992-03-09 2002-02-26 Protein Design Labs, Inc. Antibodies with increased binding affinity
US5714350A (en) 1992-03-09 1998-02-03 Protein Design Labs, Inc. Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
US5424073A (en) 1992-03-23 1995-06-13 Georgetown University Liposome encapsulated taxol and a method of using the same
WO1993021232A1 (en) 1992-04-10 1993-10-28 Research Development Foundation IMMUNOTOXINS DIRECTED AGAINST c-erbB-2 (HER-2/neu) RELATED SURFACE ANTIGENS
US5362831A (en) 1992-06-19 1994-11-08 Farmitalia Carlo Erba S.R.L. Polymer-bound paclitaxel derivatives
US5641803A (en) 1992-08-03 1997-06-24 Bristol-Myers Squibb Company Methods for administration of taxol
EP0592106A1 (en) 1992-09-09 1994-04-13 Immunogen Inc Resurfacing of rodent antibodies
US5869680A (en) 1992-10-05 1999-02-09 Rhone-Poulenc Rorer, S.A. Process for preparing taxane derivatives
US5728687A (en) 1992-11-10 1998-03-17 Rhone-Poulenc Rorer, S.A. Antitumour compositions containing taxane derivatives
US5438072A (en) 1992-12-02 1995-08-01 Rhone-Poulenc Rorer S.A. Taxoid-based compositions
US5635483A (en) 1992-12-03 1997-06-03 Arizona Board Of Regents Acting On Behalf Of Arizona State University Tumor inhibiting tetrapeptide bearing modified phenethyl amides
US6034065A (en) 1992-12-03 2000-03-07 Arizona Board Of Regents Elucidation and synthesis of antineoplastic tetrapeptide phenethylamides of dolastatin 10
US5380751A (en) 1992-12-04 1995-01-10 Bristol-Myers Squibb Company 6,7-modified paclitaxels
US5410024A (en) 1993-01-21 1995-04-25 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide amides
US5780588A (en) 1993-01-26 1998-07-14 Arizona Board Of Regents Elucidation and synthesis of selected pentapeptides
WO1994016729A1 (en) 1993-01-28 1994-08-04 Neorx Corporation Targeted nitric oxide pathway or nitric oxide synthase modulation
US5433364A (en) 1993-02-19 1995-07-18 Dynetics Engineering Corporation Card package production system with burster and carrier verification apparatus
US6214345B1 (en) 1993-05-14 2001-04-10 Bristol-Myers Squibb Co. Lysosomal enzyme-cleavable antitumor drug conjugates
US5415869A (en) 1993-11-12 1995-05-16 The Research Foundation Of State University Of New York Taxol formulation
WO1995015770A1 (en) 1993-12-09 1995-06-15 Neorx Corporation Pretargeting methods and compounds
US5773001A (en) 1994-06-03 1998-06-30 American Cyanamid Company Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
US5739116A (en) 1994-06-03 1998-04-14 American Cyanamid Company Enediyne derivatives useful for the synthesis of conjugates of methyltrithio antitumor agents
US5767285A (en) 1994-06-03 1998-06-16 American Cyanamid Company Linkers useful for the synthesis of conjugates of methyltrithio antitumor agents
US5877296A (en) 1994-06-03 1999-03-02 American Cyanamid Company Process for preparing conjugates of methyltrithio antitumor agents
US5665671A (en) 1994-07-21 1997-09-09 Hydrogeo North America Compound to defend plants from vegetal parasites
US5665860A (en) 1994-08-01 1997-09-09 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory peptide amides
US5530097A (en) 1994-08-01 1996-06-25 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory peptide amides
US5521284A (en) 1994-08-01 1996-05-28 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide amides and esters
US5504191A (en) 1994-08-01 1996-04-02 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide methyl esters
US5554725A (en) 1994-09-14 1996-09-10 Arizona Board Of Regents Acting On Behalf Of Arizona State University Synthesis of dolastatin 15
US5599902A (en) 1994-11-10 1997-02-04 Arizona Board Of Regents Acting On Behalf Of Arizona State University Cancer inhibitory peptides
US5663149A (en) 1994-12-13 1997-09-02 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide heterocyclic and halophenyl amides
US5840929A (en) 1995-04-14 1998-11-24 Bristol-Myers Squibb Company C4 methoxy ether derivatives of paclitaxel
US5705503A (en) 1995-05-25 1998-01-06 Goodall; Brian Leslie Addition polymers of polycycloolefins containing functional substituents
US5712374A (en) 1995-06-07 1998-01-27 American Cyanamid Company Method for the preparation of substantiallly monomeric calicheamicin derivative/carrier conjugates
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
US5824805A (en) 1995-12-22 1998-10-20 King; Dalton Branched hydrazone linkers
WO1997029131A1 (en) 1996-02-09 1997-08-14 Basf Aktiengesellschaft HUMAN ANTIBODIES THAT BIND HUMAN TNF$g(a)
US6090382A (en) 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
WO1997031655A2 (en) 1996-02-27 1997-09-04 Fondazione Centro San Raffaele Del Monte Tabor Modified cytokines for therapeutic use
US5714352A (en) 1996-03-20 1998-02-03 Xenotech Incorporated Directed switch-mediated DNA recombination
US5821263A (en) 1996-08-26 1998-10-13 Bristol-Myers Squibb Company Sulfenamide taxane derivatives
US5773464A (en) 1996-09-30 1998-06-30 Bristol-Myers Squibb Company C-10 epoxy taxanes
WO1998022451A1 (en) 1996-11-19 1998-05-28 Daiichi Pharmaceutical Co., Ltd. Taxol derivatives
WO1998028288A1 (en) 1996-12-24 1998-07-02 Bristol-Myers Squibb Company 6-thio-substituted paclitaxels
WO1998035704A1 (en) 1997-02-13 1998-08-20 Bone Care International, Inc. Targeted therapeutic delivery of vitamin d compounds
US6239104B1 (en) 1997-02-25 2001-05-29 Arizona Board Of Regents Isolation and structural elucidation of the cytostatic linear and cyclo-depsipeptides dolastatin 16, dolastatin 17, and dolastatin 18
WO1999009021A1 (en) 1997-08-18 1999-02-25 Florida State University Process for selective derivatization of taxanes
WO1999014209A1 (en) 1997-09-17 1999-03-25 Kabushiki Kaisha Yakult Honsha New taxane derivatives
WO1999018113A1 (en) 1997-10-08 1999-04-15 Bio Research Corporation Of Yokohama Taxoid derivatives and process for producing the same
WO1999019500A1 (en) 1997-10-10 1999-04-22 The Government Of The United States Of America, Represented By The Secretary, Department Of Healt H And Human Services Complex of biotinylated viral vector and ligand for targeted gene delivery
WO1999054342A1 (en) 1998-04-20 1999-10-28 Pablo Umana Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
US6410319B1 (en) 1998-10-20 2002-06-25 City Of Hope CD20-specific redirected T cells and their use in cellular immunotherapy of CD20+ malignancies
EP1176195A1 (en) 1999-04-09 2002-01-30 Kyowa Hakko Kogyo Co., Ltd. Method for controlling the activity of immunologically functional molecule
WO2001009785A1 (en) 1999-07-31 2001-02-08 Kyu Jin Park Study method and apparatus using digital audio and caption data
US6323315B1 (en) 1999-09-10 2001-11-27 Basf Aktiengesellschaft Dolastatin peptides
WO2001090198A1 (en) 2000-05-24 2001-11-29 Ludwig Institute For Cancer Research Multicomponent conjugates which bind to target molecules and stimulate cell lysis
US20020137134A1 (en) 2000-06-28 2002-09-26 Gerngross Tillman U. Methods for producing modified glycoproteins
US20040018590A1 (en) 2000-06-28 2004-01-29 Gerngross Tillman U. Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
US7223837B2 (en) 2001-03-23 2007-05-29 Syntarga B.V. Elongated and multiple spacers in activatible prodrugs
WO2002088172A2 (en) 2001-04-30 2002-11-07 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
US20050009751A1 (en) 2001-04-30 2005-01-13 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
US20030083263A1 (en) 2001-04-30 2003-05-01 Svetlana Doronina Pentapeptide compounds and uses related thereto
US6441163B1 (en) 2001-05-31 2002-08-27 Immunogen, Inc. Methods for preparation of cytotoxic conjugates of maytansinoids and cell binding agents
WO2003016466A2 (en) 2001-08-17 2003-02-27 Eli Lilly And Company ANTI-Aβ ANTIBODIES
WO2003035835A2 (en) 2001-10-25 2003-05-01 Genentech, Inc. Glycoprotein compositions
WO2003093793A2 (en) 2002-05-01 2003-11-13 Trellis Bioscience, Inc. Binary or polynary targeting and uses thereof
WO2004010957A2 (en) 2002-07-31 2004-02-05 Seattle Genetics, Inc. Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
US20060074008A1 (en) 2002-07-31 2006-04-06 Senter Peter D Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
US6913748B2 (en) 2002-08-16 2005-07-05 Immunogen, Inc. Cross-linkers with high reactivity and solubility and their use in the preparation of conjugates for targeted delivery of small molecule drugs
US20050271615A1 (en) 2002-08-30 2005-12-08 Doron Shabat Self-immolative dendrimers releasing many active moieties upon a single activating event
US20060116422A1 (en) 2002-11-14 2006-06-01 De Groot Franciscus Marinus H Prodrugs built as multiple self-elimination-release spacers
WO2004050016A2 (en) 2002-11-27 2004-06-17 The Regents Of The University Of California Delivery of pharmaceutical agents via the human insulin receptor
US8535678B2 (en) 2003-02-20 2013-09-17 Seattle Genetics, Inc. Anti-CD70 antibody-drug conjugates and their use for the treatment of cancer and immune disorders
US20050042664A1 (en) 2003-08-22 2005-02-24 Medimmune, Inc. Humanization of antibodies
US20050238649A1 (en) 2003-11-06 2005-10-27 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
WO2005081898A2 (en) 2004-02-20 2005-09-09 The Trustees Of The University Of Pennsylvania Binding peptidomimetics and uses of the same
US7989434B2 (en) 2004-02-23 2011-08-02 Seattle Genetics, Inc. Heterocyclic self-immolative linkers and conjugates
US20070173497A1 (en) 2004-03-01 2007-07-26 Spirogen Limited 11-Hydroxy-5h-pyrrolo[2,1-c][1,4] benzodiazepin-5-one derivatives as key intermediates for the preparation of c2 substituted pyrrolobenzodiazepines
US7741319B2 (en) 2004-03-01 2010-06-22 Spirogen Limited 11-hydroxy-5h-pyrrolo[2,1-c][1,4] benzodiazepin-5-one derivatives as key intermediates for the preparation of c2 substituted pyrrolobenzodiazepines
WO2005100584A2 (en) 2004-04-15 2005-10-27 Glycofi, Inc. Production of galactosylated glycoproteins in lower eukaryotes
US20060024317A1 (en) 2004-05-19 2006-02-02 Medarex, Inc Chemical linkers and conjugates thereof
US8142784B2 (en) 2004-06-01 2012-03-27 Genentech, Inc. Antibody-drug conjugates and methods
WO2006083562A2 (en) 2005-02-02 2006-08-10 Awdalla, Essam, T. Method and multicomponent conjugates for treating cancer
WO2006089668A1 (en) 2005-02-23 2006-08-31 Ludwig-Maximilians-Universität Transport of nano- and macromolecular structures into cytoplasm and nucleus of cells
WO2006111759A1 (en) 2005-04-21 2006-10-26 Spirogen Limited Pyrrolobenzodiazepines
US20120107332A1 (en) 2005-07-18 2012-05-03 Seattle Genetics, Inc. Beta-Glucuronide-Linker Drug Conjugates
US8568728B2 (en) 2005-07-18 2013-10-29 Seattle Genetics, Inc. Beta-glucuronide-linker drug conjugates
US20090318668A1 (en) 2006-02-02 2009-12-24 Patrick Henry Beusker Water-Soluble CC-1065 Analogs and Their Conjugates
WO2007150020A1 (en) 2006-06-23 2007-12-27 Simon Paul M Targeted immune conjugates
US7598028B2 (en) 2006-11-28 2009-10-06 The Regents Of The University Of Michigan Compositions and methods for detecting and treating prostate disorders
WO2008097866A2 (en) 2007-02-02 2008-08-14 Baylor Research Institute Vaccines based on targeting antigen to dcir expressed an antigen-presenting cells
WO2008097870A2 (en) 2007-02-02 2008-08-14 Baylor Research Institute Agents that engage antigen - presenting cells through dendritic cell asialoglycoprotein receptor (dc- asgpr)
WO2008100624A2 (en) 2007-02-16 2008-08-21 Merrimack Pharmaceuticals, Inc. Antibodies against erbb3 and uses thereof
WO2008103947A2 (en) 2007-02-23 2008-08-28 Baylor Research Institute Activation of human antigen-presenting cells through clec-6
WO2008103953A2 (en) 2007-02-23 2008-08-28 Baylor Research Institute Activation of human antigen-presenting cells through dendritic cell lectin-like oxidized ldl receptor-1 (lox-1)
US8389282B2 (en) 2007-03-30 2013-03-05 Memorial Sloan-Kettering Cancer Center Constitutive expression of costimulatory ligands on adoptively transferred T lymphocytes
WO2008135237A1 (en) 2007-05-03 2008-11-13 Medizinische Universität Innsbruck Complement factor h-derived short consensus repeat-antibody constructs
US8399512B2 (en) 2007-11-28 2013-03-19 Mersana Therapeutics, Inc. Biocompatible biodegradable fumagillin analog conjugates
US9105939B2 (en) 2008-03-10 2015-08-11 Nissan Motor Co., Ltd. Battery with battery electrode and method of manufacturing same
US8236319B2 (en) 2008-04-30 2012-08-07 Immunogen, Inc. Cross-linkers and their uses
US8822647B2 (en) 2008-08-26 2014-09-02 City Of Hope Method and compositions using a chimeric antigen receptor for enhanced anti-tumor effector functioning of T cells
US20100152725A1 (en) 2008-12-12 2010-06-17 Angiodynamics, Inc. Method and system for tissue treatment utilizing irreversible electroporation and thermal track coagulation
US8309093B2 (en) 2009-03-06 2012-11-13 Agensys, Inc. Antibody drug conjugates (ADC) that bind to 24P4C12 proteins
WO2010111198A1 (en) 2009-03-23 2010-09-30 Quark Pharmaceuticals, Inc. Compounds compositions and methods of treating cancer and fibrotic diseases
US8524214B2 (en) 2009-05-28 2013-09-03 Mersana Therapeutics, Inc. Polyal drug conjugates comprising variable rate-releasing linkers
WO2011000370A1 (en) 2009-06-30 2011-01-06 Röhm Gmbh Drilling device
US20110076232A1 (en) 2009-09-29 2011-03-31 Ludwig Institute For Cancer Research Specific binding proteins and uses thereof
WO2011057216A1 (en) 2009-11-06 2011-05-12 The Pennsylvania State Research Foundation Bioconjugation of calcium phosphosilicate nanoparticles for selective targeting of cells in vivo
WO2011058321A1 (en) 2009-11-11 2011-05-19 King's College Hospital Nhs Foundation Trust Conjugate molecule
US20140328750A1 (en) * 2010-03-04 2014-11-06 Macrogenics, Inc. Antibodies Reactive with B7-H3 and Uses Thereof
WO2011109400A2 (en) * 2010-03-04 2011-09-09 Macrogenics,Inc. Antibodies reactive with b7-h3, immunologically active fragments thereof and uses thereof
US8349308B2 (en) 2010-03-26 2013-01-08 Mersana Therapeutics, Inc. Modified polymers for delivery of polynucleotides, method of manufacture, and methods of use thereof
US20130028917A1 (en) 2010-04-15 2013-01-31 Spirogen Developments Sàrl Pyrrolobenzodiazepines and conjugates thereof
US20130028919A1 (en) 2010-04-15 2013-01-31 Spirogen Developments Sàrl Targeted pyrrolobenzodiazapine conjugates
WO2011130598A1 (en) 2010-04-15 2011-10-20 Spirogen Limited Pyrrolobenzodiazepines and conjugates thereof
WO2012027494A1 (en) 2010-08-24 2012-03-01 Regents Of The University Of Minnesota Bispecific targeting reagents
US8975071B1 (en) 2010-12-09 2015-03-10 The Trustees Of The University Of Pennsylvania Compositions for treatment of cancer
US8916381B1 (en) 2010-12-09 2014-12-23 The Trustees Of The University Of Pennyslvania Methods for treatment of cancer
US8911993B2 (en) 2010-12-09 2014-12-16 The Trustees Of The University Of Pennsylvania Compositions for treatment of cancer
US8906682B2 (en) 2010-12-09 2014-12-09 The Trustees Of The University Of Pennsylvania Methods for treatment of cancer
US20130224228A1 (en) 2011-12-05 2013-08-29 Igenica, Inc. Antibody-Drug Conjugates and Related Compounds, Compositions, and Methods
US20130189218A1 (en) 2011-12-23 2013-07-25 Mersana Therapeutics, Inc. Pharmaceutical formulations for fumagillin derivative-phf conjugates
US20130303509A1 (en) 2012-05-11 2013-11-14 Abbvie Inc. Nampt inhibitors
WO2013173337A2 (en) 2012-05-15 2013-11-21 Seattle Genetics, Inc. Self-stabilizing linker conjugates
US20130309256A1 (en) 2012-05-15 2013-11-21 Seattle Genetics, Inc. Self-stabilizing linker conjugates
US20140017265A1 (en) 2012-07-05 2014-01-16 Mersana Therapeutics, Inc. Terminally Modified Polymers and Conjugates Thereof
WO2014093394A1 (en) 2012-12-10 2014-06-19 Mersana Therapeutics, Inc. Protein-polymer-drug conjugates
WO2014093379A1 (en) 2012-12-10 2014-06-19 Mersana Therapeutics, Inc. Auristatin compounds and conjugates thereof
WO2014093640A1 (en) 2012-12-12 2014-06-19 Mersana Therapeutics,Inc. Hydroxy-polmer-drug-protein conjugates
US9109630B2 (en) 2013-01-15 2015-08-18 Jtekt Corporation Rolling bearing unit
US20140322275A1 (en) 2013-02-20 2014-10-30 Jennifer Brogdon TREATMENT OF CANCER USING HUMANIZED ANTI-EGFRvIII CHIMERIC ANTIGEN RECEPTOR
US20140286968A1 (en) 2013-03-15 2014-09-25 Abbvie Inc. Antibody drug conjugate (adc) purification
US9401234B2 (en) 2013-03-22 2016-07-26 Polytronics Technology Corp. Over-current protection device
US9618978B2 (en) 2013-10-03 2017-04-11 Acer Incorporated Methods for controlling a touch panel and portable computers using the same
US20160158377A1 (en) 2014-12-09 2016-06-09 Abbvie Inc. BCL-XL Inhibitory Compounds and Antibody Drug Conjugates Including the Same
US9816280B1 (en) 2016-11-02 2017-11-14 Matthew Reitnauer Portable floor

Non-Patent Citations (167)

* Cited by examiner, † Cited by third party
Title
"Cancer Therapy with Radiolabeled Antibodies", 1995, CRC PRESS
"Current Protocols in Molecular Biology", 1993, JOHN WILEY &SONS
"Monoclonal Antibodies For Cancer Detection And Therapy,", 1985, ACADEMIC PRESS, article "Analysis, Results, and Future Prospective of the Therapeutic Use of Radiolabeled Antibody In Cancer Therapy"
AKIKO NAGASE-ZEMBUTSU ET AL: "Development of DS-5573a: A novel afucosylated mAb directed at B7-H3 with potent antitumor activity", CANCER SCIENCE, vol. 107, no. 5, 26 April 2016 (2016-04-26), JP, pages 674 - 681, XP055396124, ISSN: 1347-9032, DOI: 10.1111/cas.12915 *
AMIR ET AL., ANGEW. CHEM. INT. ED., vol. 42, 2003, pages 4494 - 4499
AMON ET AL.: "Monoclonal Antibodies And Cancer Therapy", 1985, ALAN R. LISS, INC., article "Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy"
AMSBERRY ET AL., J. ORG. CHEM., vol. 55, 1990, pages 5867
AMUNDSON ET AL., CANCER RESEARCH, vol. 60, 2000, pages 6101 - 6110
ANONYMOUS: "Macrogenics - Corporate Fact Sheet", 26 May 2016 (2016-05-26), pages 1 - 2, XP055396257, Retrieved from the Internet <URL:https://www.macrogenics.com/wp-content/uploads/2016/05/MacroGenics_Corporate_Factsheet.pdf> [retrieved on 20170804] *
AUBEL ET AL., PROC. NATL. ACAD. SCI., USA, vol. 88, 1991, pages 906
AXUP ET AL., PROC NATL ACAD SCI, vol. 109, 2003, pages 16101 - 16106
BADESCU ET AL., BIOCONJUGATE CHEM., vol. 25, 2014, pages 1124 - 1136
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 426
BOGER, PURE AND APPL CHEM, vol. 65, no. 6, 1993, pages 1123
BOGER; JOHNSON, PNAS USA, vol. 92, 1995, pages 3642
BROWN ET AL., J. AM. CHEM. SOC., vol. 86, 1968, pages 397
BURKE ET AL., BIOCONJUGATE CHEM, vol. 20, 2009, pages 1242 - 1250
CAMPOS, CYTOMETRY A, vol. 69, no. 6, 2006, pages 515 - 523
CARTER ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 89, 1992, pages 4285
CHAPOVAL ET AL., NAT. IMMUNOL., vol. 2, 2001, pages 269 - 274
CHARI ET AL., CANCER RES, vol. 52, 1992, pages 127
CHARI ET AL., CANCER RESEARCH, vol. 52, 1992, pages 127 - 131
CHARI, ACC CHEM RES, vol. 41, 2008, pages 98 - 107
CHOTHIA ET AL., NATURE, vol. 342, 1989, pages 877 - 883
CHOTHIA; LESK, J. MOL. BIOL., vol. 196, 1987, pages 901
CHOTHIA; LESK, J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
CO, M.S. ET AL., MOL. IMMUNOL., vol. 30, 1993, pages 1361 - 1367
CROOKE ST., BIOCHIM. BIOPHYS. ACTA, vol. 1489, 1999, pages 30 - 42
D. LOO ET AL: "Development of an Fc-Enhanced Anti-B7-H3 Monoclonal Antibody with Potent Antitumor Activity", CLINICAL CANCER RESEARCH, vol. 18, no. 14, 15 July 2012 (2012-07-15), pages 3834 - 3845, XP055092714, ISSN: 1078-0432, DOI: 10.1158/1078-0432.CCR-12-0715 *
DAMLE ET AL., CURR OPIN PHARMACOL, vol. 3, 2003, pages 386
DERYK LOO ET AL: "Abstract 1201: Anti-B7-H3 antibody-drug conjugates as potential therapeutics for solid cancer", CANCER RESEARCH, 20 April 2016 (2016-04-20), pages 1201 - 1201, XP055396737, Retrieved from the Internet <URL:http://files.shareholder.com/downloads/AMDA-278VRP/0x0x886237/E3742E18-80FE-4E9D-BA3B-FDA5E0B5EA32/MacroGenics_AACR_2016_-_Anti-B7-H3_Antibody-drug_Conjugates.pdf> [retrieved on 20170807], DOI: 10.1158/1538-7445.AM2016-1201 *
DICK ET AL., BIOTECHNOL. BIOENG., vol. 100, 2008, pages 1132
DICK ET AL., BIOTECHNOL. BIOENG., vol. 97, 2007, pages 544
DORONINA ET AL., NAT. BIOTECHNOL., vol. 21, 2003, pages 778 - 784
DORONINA ET AL.: "Development of potent and highly efficacious monoclonal antibody auristatin conjugates for cancer therapy", NAT. BIOTECHNOL., vol. 21, no. 7, 2003, pages 778 - 784
DORR RT ET AL., CANCER RES., vol. 45, 1985, pages 3510
DUBOWCHIK ET AL., BIOORG. MED. CHEM. LETT., vol. 8, 1998, pages 3341 - 3346
DUBOWCHIK ET AL., J. ORG. CHEM., vol. 67, 1998, pages 1866 - 1872
DUBOWCHIK; WALKER, PHARM. THERAPEUTICS, vol. 83, 1999, pages 67 - 123
DUCRY ET AL., BIOCONJUGATE CHEM., vol. 21, 2010, pages 5 - 13
E. PICARDA ET AL: "Molecular Pathways: Targeting B7-H3 (CD276) for Human Cancer Immunotherapy", CLINICAL CANCER RESEARCH, vol. 22, no. 14, 20 May 2016 (2016-05-20), US, pages 3425 - 3431, XP055396638, ISSN: 1078-0432, DOI: 10.1158/1078-0432.CCR-15-2428 *
FIRE ET AL., NATURE, vol. 391, 1998, pages 806 - 811
FRANCISCO ET AL., BLOOD, vol. 102, 2003, pages 1458 - 1465
FRANCISCO ET AL.: "cAClO-vcMMAE, an anti-CD30-monomethylauristatin E conjugate with potent and selective antitumor activity", BLOOD, vol. 102, 2003, pages 1458 - 1465
G. R. PETTIT, PROG. CHEM. ORG. NAT. PROD, vol. 70, 1997, pages 1 - 79
GELBOIN ET AL., SCIENCE, vol. 167, 1970, pages 205 - 207
GILLIES ET AL., J. IMMUNOL. METHODS, vol. 125, 1989, pages 191 - 202
GOLDSPIEL ET AL., CLINICAL PHARMACY, vol. 12, 1993, pages 488 - 505
GOLDSTEIN ET AL., CELL DEATH AND DIFFERENTIATION, vol. 12, 2005, pages 453 - 462
GROOT ET AL., ANGEW. CHEM. INT. ED., vol. 42, 2003, pages 4490 - 4494
HAMBLETT ET AL.: "Effects of Drug Loading on the Antitumor Activity of a Monoclonal Antibody Drug Conjugate", CLIN. CANCER RES., vol. 10, 2004, pages 7063 - 7070, XP002726047, DOI: doi:10.1158/1078-0432.CCR-04-0789
HASHIGUCHI ET AL., PROC. NAT'L. ACAD. SCI. U.S.A., vol. 105, no. 30, 2008, pages 10495 - 10500
HAY ET AL., BIOORG. MED. CHEM. LETT., vol. 9, 1999, pages 2237
HELLSTROM ET AL.: "Controlled Drug Delivery", 1987, MARCEL DEKKER, INC., article "Antibodies For Drug Delivery"
HINMAN ET AL., CANCER RESEARCH, vol. 53, 1993, pages 3336 - 3342
HOLLANDER ET AL., BIOCONJUGATE CHEM, vol. 19, 2008, pages 358 - 361
HOLLIGER, P. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448
HUSTON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879 - 5883
JEFFREY ET AL., BIOCONJUG. CHEM., vol. 17, 2006, pages 831 - 840
JEFFREY ET AL., BIOORG. MED. CHEM. LETT., vol. 17, 2007, pages 2278 - 2280
JIANG ET AL., J. AM . CHEM. SOC., vol. 127, 2005, pages 11254 - 11255
JIANG ET AL., J. AM. CHEM. SOC., vol. 127, 2005, pages 11254 - 11255
JOHNNSON, B. ET AL., ANAL. BIOCHEM., vol. 198, 1991, pages 268 - 277
JOHNSON ET AL., ANTICANCER RES., vol. 15, pages 1387 - 93
JOHNSSON, B. ET AL., J. MOL. RECOGNIT., vol. 8, 1995, pages 125 - 131
JONES ET AL., NATURE, vol. 321, 1986, pages 522
JONSSON, U. ET AL., ANN. BIOL. CLIN., vol. 51, 1993, pages 19 - 26
JONSSON, U. ET AL., BIOTECHNIQUES, vol. 11, 1991, pages 620 - 627
KABAT ET AL., ANN. NY ACAD, SCI., vol. 190, 1971, pages 382 - 391
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1987, NATIONAL INSTITUTES OF HEALTH
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", U.S. DEPT. HEALTH, 1983
KABAT, E.A. ET AL.: "Sequences of Proteins of Immunological Interest", 1991, U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES, NIH PUBLICATION, article "Sequences of Proteins of Immunological Interest"
KAMIJO ET AL., ORG. LETT., vol. 13, 2011, pages 5928 - 5931
KANEKO ET AL., BIOCONJUGATE CHEM., vol. 2, 1991, pages 133 - 41
KENNEDY KA ET AL., CANCER RES., vol. 45, 1985, pages 3541
KING ET AL., J MED CHEM, vol. 45, 2002, pages 4336 - 4343
KING ET AL., TETRAHEDRON LETTERS, vol. 43, 2002, pages 1987 - 1990
KINGSBURY ET AL., J. MED. CHEM., vol. 27, 1984, pages 1447
KIRKIN ET AL., BIOCHIMICA BIOPHYSICA ACTA, vol. 1644, 2004, pages 229 - 249
KITSON ET AL., CROS/CMOS - CHEMICA OGGI - CHEMISTRY TODAY, vol. 31, no. 4, 2013, pages 30 - 36
KONTERMANN; DUBEL: "Antibody Engineering", 2001, SPRINGER-VERLAG, pages: 790
KRIEGLER: "A Laboratory Manual", 1990, STOCKTON PRESS, article "Gene Transfer and Expression"
KUPCHAN ET AL., J. AM. CHEM. SOC., vol. 94, 1972, pages 1354 - 1356
KUPCHAN ET AL., J. MED. CHEM., vol. 21, 1978, pages 31 - 37
LAU ET AL., BIOORG-MED-CHEM., vol. 3, no. 10, 1995, pages 1299 - 1304
LAU ET AL., BIOORG-MED-CHEM., vol. 3, no. 10, 1995, pages 1305 - 12
LEBEL ET AL., ORG. LETT., vol. 7, 2005, pages 4107 - 4110
LEE; MARGOLIN, CANCERS, vol. 3, 2011, pages 3856
LEVY ET AL., PROC. NAT. ACAD. SCI. USA, vol. 62, 1969, pages 357 - 361
LIU ET AL., J. BIOL. CHEM., vol. 286, 2011, pages 11211
LIU ET AL., JBC, vol. 286, 2011, pages 11211
LODE ET AL., CANCER RESEARCH, vol. 58, 1998, pages 2925 - 2928
LOOS ET AL., BMC CANCER, vol. 9, 2009, pages 463
LYON ET AL., NAT. BIOTECHNOL., vol. 32, 2014, pages 1059 - 1062
MACCALLUM, J. MOL. BIOL., vol. 262, no. 5, 1996, pages 732 - 745
MAHIUDDIN AHMED ET AL: "Humanized Affinity-matured Monoclonal Antibody 8H9 Has Potent Antitumor Activity and Binds to FG Loop of Tumor Antigen B7-H3", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 290, no. 50, 11 December 2015 (2015-12-11), US, pages 30018 - 30029, XP055396132, ISSN: 0021-9258, DOI: 10.1074/jbc.M115.679852 *
MAY, TIBTECH, vol. 11, no. 5, 1993, pages 155 - 215
MORGAN; ANDERSON, ANN. REV. BIOCHEM., vol. 62, 1993, pages 191 - 217
MORRISON ET AL., PROC. NATL. ACAD. SCI., vol. 81, 1984, pages 851 - 855
MORRISON, SCIENCE, vol. 229, 1985, pages 1202
MULLIGAN, SCIENCE, vol. 260, 1993, pages 926 - 932
NAGAMURA; SAITO, CHEMISTRY OF HETEROCYCLIC COMPOUNDS, vol. 34, no. 12, 1998
NEUBERGER ET AL., NATURE, vol. 312, 1984, pages 604 - 608
NEVILLE ET AL., BIOL. CHEM., vol. 264, 1989, pages 14653 - 14661
NOLTING, ANTIBODY-DRUG CONJUGATES, METHODS IN MOLECULAR BIOLOGY, vol. 1045, 2013, pages 71 - 100
NOLTING: "In: Antibody-Drug Conjugates: Methods in Molecular Biology", vol. 1045, 2013, SPRINGER SCIENCE & BUSINESS MEDICA, LLC, article "Linker Technology in Antibody-Drug Conjugates", pages: 71 - 100
NYGREN ET AL., FRONT BIOSCI., vol. 3, 2011, pages 989 - 993
OI ET AL., BIOTECHNIQUES, vol. 4, 1986, pages 214
PADLAN, MOLECULAR IMMUNOLOGY, vol. 28, no. 4/5, 1991, pages 489 - 498
PETTIT ET AL., J. AM. CHEM. SOC., vol. 98, 1976, pages 4677
POLJAK, R.J. ET AL., STRUCTURE, vol. 2, 1994, pages 1121 - 1123
POWELL ET AL., J. NAT. PROD., vol. 46, 1983, pages 660 - 666
PRASAD ET AL., J. IMMUNOL., vol. 173, 2004, pages 2500 - 2506
PRESTA ET AL., J. IMMUNOL., vol. 151, 1993, pages 2623
R. JEFFERIS, BIOTECHNOL. PROG., vol. 21, 2005, pages 11 - 16
R.J. KAUFMAN; P.A. SHARP, MOL. BIOL., vol. 159, 1982, pages 601 - 621
REMILLARD ET AL., SCIENCE, vol. 189, 1975, pages 1002 - 1005
RIECHMANN ET AL., NATURE, vol. 332, 1988, pages 323
ROBERTS ET AL., J. ORG. CHEM., vol. 59, 1994, pages 6464 - 6469
ROCKVILLE ET AL: "MacroGenics Presents Data from Five Preclinical Programs at AACR Annual Meeting 2016", 19 April 2016 (2016-04-19), XP055396285, Retrieved from the Internet <URL:http://ir.macrogenics.com/common/download/download.cfm?companyid=AMDA-278VRP&fileid=886563&filekey=03D3915B-D68D-4906-B67E-BB61941CF359&filename=965754.pdf> [retrieved on 20170807] *
RODRIGUES ET AL., CHEMISTRY BIOLOGY, vol. 2, 1995, pages 223
ROGUSKA ET AL., PNAS, vol. 91, 1994, pages 969 - 973
ROTH ET AL., CANCER RES., vol. 67, no. 16, 2007, pages 7893 - 7900
RUVKUN, SCIENCE, vol. 2294, 2001, pages 797
SAKAI ET AL., J. NAT. PROD., vol. 51, 1988, pages 845 - 850
SELLEI C ET AL., CANCER CHEMOTHER REP., vol. 53, 1969, pages 377
SHAMIS ET AL., J. AM. CHEM. SOC., vol. 126, 2004, pages 1726 - 1731
SHIELDS, R. L. ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 26733 - 26740
SIMS ET AL., J. IMMUNOL., vol. 151, 1993, pages 2296
STEINBERGER ET AL., J. IMMUNOL., vol. 172, no. 4, 2004, pages 2352 - 2359
STORM ET AL., J. AMER. CHEM. SOC., vol. 94, 1972, pages 5815
STUDNICKA ET AL., PROTEIN ENGINEERING, vol. 7, no. 6, 1994, pages 805 - 814
SUH ET AL., NAT. IMMUNOL., vol. 4, 2003, pages 899 - 906
SUN ET AL., BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 12, 2002, pages 2213 - 2215
SUN ET AL., BIOORGANIC & MEDICINAL CHEMISTRY, vol. 11, 2003, pages 1761 - 1768
SUN ET AL., LUNG CANCER, vol. 53, no. 2, 2006, pages 143 - 151
SUTHERLAND ET AL., BLOOD, vol. 122, no. 8, 2013, pages 1455
SUWANBORIRUX ET AL., EXPERIENTIA, vol. 46, 1990, pages 117 - 120
TAKEDA ET AL., NATURE, vol. 314, 1985, pages 452 - 454
TAO ET AL., ACS MED. CHEM. LETT., vol. 5, 2014, pages 1088 - 1093
THORPE ET AL., CANCER RES., vol. 47, 1987, pages 5924 - 5931
THORPE ET AL., IMMUNOL. REV., vol. 62, 1982, pages 119 - 158
THORPE ET AL.: "Monoclonal Antibodies '84: Biological And Clinical Applications", 1985, article "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review"
TIAN ET AL., PROC NATL ACAD SCI, vol. 111, 2014, pages 1776 - 1771
TOLSTOSHEV, ANN. REV. PHARMACOL. TOXICOL., vol. 32, 1993, pages 573 - 596
TUMEY ET AL., BIOCONJUGATE CHEM., vol. 25, 2014, pages 1871 - 1880
UMANA ET AL., NAT. BIOTECH., vol. 17, 1999, pages 176 - 180
URLAUB; CHASIN, PROC. NATL. ACAD. SCI. USA, vol. 77, 1980, pages 4216 - 4220
VERHOEYEN ET AL., SCIENCE, vol. 239, 1988, pages 1534
WALKER ET AL., BIOORG. MED. CHEM. LETT., vol. 12, 2002, pages 217 - 219
WALKER ET AL., BIOORG. MED. CHEM. LETT., vol. 14, 2004, pages 4323 - 4327
WALLICK, S.C. ET AL., EXP. MED., vol. 168, 1988, pages 1099 - 1109
WANG ET AL., EUR. J. IMMUNOL., vol. 35, 2005, pages 428 - 438
WANI ET AL., J. CHEM. SOC. CHEM. COMMUN., vol. 390, 1973
WARD ET AL., NATURE, vol. 341, 1989, pages 544 - 546
WAWRZYNCZAK ET AL.: "Immunoconjugates: Antibody Conjugates in Radioimagery and Therapy of Cancer", 1987, OXFORD U. PRESS
WINNAKE: "From Genes to Clones", 1987, VERLAGSGESELLSCHAFT
WRIGHT, A. ET AL., EMBO J., vol. 10, 1991, pages 2717 - 2723
WU ET AL., WORLD J. GASTROENTEROL., vol. 12, no. 3, 2006, pages 457 - 459
WU; WU, BIOTHERAPY, vol. 3, 1991, pages 87 - 95
YAMAMOTO ET AL., HETEROCYCLES, vol. 47, 1998, pages 765 - 780
YAMATO ET AL., BR. J. CANCER, vol. 101, no. 10, 2009, pages 1709 - 1716
YANG ET AL., ORG. LETT., vol. 15, 2013, pages 690 - 693
ZAMORE PD., SCIENCE, vol. 296, 2002, pages 1265 - 1269
ZELEZNICK ET AL., PROC. SOC. EXP. BIOL. MED., vol. 130, 1969, pages 126 - 128
ZHANG, NATURE REVIEWS/DRUG DISCOVERY, vol. 1, 2002, pages 101
ZHAO ET AL., J. MED. CHEM., vol. 54, 2011, pages 3606 - 3623

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10640563B2 (en) 2016-06-08 2020-05-05 Abbvie Inc. Anti-B7-H3 antibodies and antibody drug conjugates
EP3835322A3 (en) * 2016-06-08 2021-10-06 AbbVie Inc. Anti-b7-h3 antibodies and antibody drug conjugates
JP7458997B2 (en) 2018-05-29 2024-04-01 ブリストル-マイヤーズ スクイブ カンパニー Modified self-cleaving moieties for use in prodrugs and conjugates and methods of use and preparation thereof
WO2020018964A1 (en) 2018-07-20 2020-01-23 Fred Hutchinson Cancer Research Center Compositions and methods for controlled expression of antigen-specific receptors
JP2022501076A (en) * 2018-09-26 2022-01-06 フーヂョウ ティーセルテック バイオテクノロジー カンパニー リミテッドFuzhou Tcelltech Biotechnology Co., Ltd. Monoclonal antibody against B7-H3 and its use in cell therapy
JP7333104B2 (en) 2018-09-26 2023-08-24 フーヂョウ ティーセルテック バイオテクノロジー カンパニー リミテッド Monoclonal antibodies against B7-H3 and their use in cell therapy
CN113507941A (en) * 2018-12-21 2021-10-15 萨普雷米科技有限公司 Bioactive molecular cluster
WO2020236817A2 (en) 2019-05-20 2020-11-26 Novartis Ag Mcl-1 inhibitor antibody-drug conjugates and methods of use
US11759527B2 (en) 2021-01-20 2023-09-19 Abbvie Inc. Anti-EGFR antibody-drug conjugates
WO2023160376A1 (en) * 2022-02-25 2023-08-31 Nanjing Probio Biotech Co., Ltd. Antibodies and variants thereof against human b7-h3

Also Published As

Publication number Publication date
JP2019521973A (en) 2019-08-08
WO2017214339A4 (en) 2018-02-08
US20200338209A1 (en) 2020-10-29
CA3027103A1 (en) 2017-12-14
MX2018015285A (en) 2019-09-18
CN109641962A (en) 2019-04-16
EP3469000A1 (en) 2019-04-17
AU2017279554A1 (en) 2019-01-03
BR112018075649A2 (en) 2019-04-09

Similar Documents

Publication Publication Date Title
US20210171637A1 (en) Anti-b7-h3 antibodies and antibody drug conjugates
US20230120736A1 (en) Anti-cd98 antibodies and antibody drug conjugates
US20200338209A1 (en) Anti-b7-h3 antibodies and antibody drug conjugates
US20200002421A1 (en) Anti-b7-h3 antibodies and antibody drug conjugates
US20230135723A1 (en) Anti-cd98 antibodies and antibody drug conjugates
US20200002432A1 (en) Anti-cd98 antibodies and antibody drug conjugates
NZ788873A (en) Anti-B7-H3 antibodies and antibody drug conjugates

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17732658

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2018564199

Country of ref document: JP

Kind code of ref document: A

Ref document number: 3027103

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112018075649

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2017279554

Country of ref document: AU

Date of ref document: 20170607

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017732658

Country of ref document: EP

Effective date: 20190108

ENP Entry into the national phase

Ref document number: 112018075649

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20181210